EP0986577A2 - Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria - Google Patents

Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Info

Publication number
EP0986577A2
EP0986577A2 EP98931278A EP98931278A EP0986577A2 EP 0986577 A2 EP0986577 A2 EP 0986577A2 EP 98931278 A EP98931278 A EP 98931278A EP 98931278 A EP98931278 A EP 98931278A EP 0986577 A2 EP0986577 A2 EP 0986577A2
Authority
EP
European Patent Office
Prior art keywords
seq
antibody
gram positive
positive bacteria
arg
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP98931278A
Other languages
German (de)
French (fr)
Other versions
EP0986577B1 (en
Inventor
Gerald W. Fischer
Richard F. Schuman
Hing Wong
Jeffrey L. Stinson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Henry M Jackson Foundation for Advancedment of Military Medicine Inc
Original Assignee
Henry M Jackson Foundation for Advancedment of Military Medicine Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Henry M Jackson Foundation for Advancedment of Military Medicine Inc filed Critical Henry M Jackson Foundation for Advancedment of Military Medicine Inc
Priority to EP10182580A priority Critical patent/EP2357198A1/en
Publication of EP0986577A2 publication Critical patent/EP0986577A2/en
Application granted granted Critical
Publication of EP0986577B1 publication Critical patent/EP0986577B1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1271Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Micrococcaceae (F), e.g. Staphylococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the invention includes monoclonal and chimeric antibodies, as well as fragments, regions and derivatives thereof.
  • invention also relates to the epitope to which the antibodies of the invention bind as well as the sequences, fragments, and regions of the epitopes. Both the antibodies and peptides that encompass the epitope, and regions and fragments thereof, may be used for diagnostic, prophylactic and therapeutic applications.
  • bacteria requires categorization, and the most fundamental categories for bacteria are their response to the Gram stain, yielding (for the most part) Gram positive bacteria and Gram negative bacteria.
  • the cells walls of Gram negative bacteria are made up of a unique outer membrane of two opposing phospholipid-protein leaflets, with an ordinary phospholipid in the inner leaflet but the extremely toxic lipopolysaccharide in the outer leaflet.
  • the cell walls of Gram positive bacteria seem much simpler in
  • Staphylococcus commonly colonize humans and animals and are an
  • Staphylococci are prevalent on the skin and mucosal linings and, accordingly, are ideally situated to produce both localized and systemic infections.
  • coagulase an enzyme that causes fibrin to coagulate and to form a
  • clot coagulase positive and coagulase negative.
  • the coagulase positive Staphylococcus species most frequently pathogenic in humans is Staphylococcus aureus.
  • S. aureus is the most virulent Staphylococcus and produces severe and
  • epidermidis is the most common coagulase negative species.
  • S. epidermidis has become a major cause of nosocomial infection in patients whose treatments include the placement of foreign objects such as cerebrospinal fluid shunts, cardiac valves, vascular catheters, joint prostheses, and other implants into the body.
  • S. epidermidis and S. aureus are common causes of post-operative wound infections and S. epidermidis has also become a common
  • Staphylococcal infections are difficult to treat for a variety of reasons.
  • these antibodies have been capsuiar polysacchahdes or surface proteins.
  • epidermidis type-specific anti-capsular antibody, suggesting that S. epidermidis and
  • S. aureas have a similar pathogenesis and opsonic requirement as other
  • the monoclonal antibody produced to the homologous strain was opsonic and
  • aureus would have opsonic activity against both. This is particularly true for
  • Staphylococci Accordingly, there is a need in the art to provide monoclonal antibodies that can bind to Staphylococcus of both coagulase types and that can enhance phagocytosis and killing of the bacteria and thereby enhance protection in vivo
  • the present invention encompasses opsonic and protective monoclonal and chimeric antibodies that bind to lipoteichoic
  • the antibodies also bind to whole bacteria and
  • the mouse monoclonal antibody has been humanized and the resulting chimeric antibody provides a previously unknown means to
  • This invention also encompasses a peptide mimic of the lipoteichoic acid epitope binding site defined by the monoclonal antibody.
  • epitope or epitope peptide mimic identifies other antibodies that may bind to the lipoteichoic acid epitope. Moreover, the epitope or epitope peptide mimic provides a valuable substrate for the generation of vaccines or other therapeutics.
  • FIG. 1 provides a schematic representation of lipoteichoic acid (LTA) in the Gram positive bacterial cell wall.
  • LTA lipoteichoic acid
  • FIG. 2 depicts antibody regions, such as the heavy chain constant region
  • C H the heavy chain variable region (V H ), the light chain constant region (C L ), and the light chain variable region (V L ).
  • Figure 3 demonstrates the enhancement of survival after administration of MAB 96-110 in a lethal neonatal model of coagulase positive staphylococcus sepsis.
  • Figure 4 demonstrates enhancement of survival in adult mice infected with coagulase negative staphylococci after administration of MAB 96-110.
  • Figure 5 (SEQ ID NOS 4 & 5, and 6 & 7, respectively) provides a list of 18
  • Figure 7 (SEQ ID NOS 44 & 45, and 46 & 47, and 48 & 49, respectively) provides a list of the 17 resulting sequences for the first experiment 15mer library
  • Figure 8 (SEQ ID NOS 50-67, respectively) provides a master list compiled from the common resulting peptide sequences from all the pannings.
  • Figure 9 sets forth a comparison of the optical density signals of each phage
  • Figure 10 shows the strategy for cloning the variable region gene fragments.
  • Figure 11 (SEQ ID NOS 68-85, respectively) lists the oligonucleotides primers
  • Figure 12 (SEQ ID NOS 86-105, respectively) provides the final consensus DNA sequence of the heavy and light chain variable regions.
  • Figure 13 demonstrates the re-amplification of the variable region gene
  • Figure 14 sets forth the plasmid map for pJRS334.
  • Figure 15 provides the results of the antibody production assay, showing that the transfection of cells with the plasmid construct results in the production of a molecule containing both human IgG and kappa domains.
  • Figure 16 provides the results of the activity assay, demonstrating that the
  • Figure 17 depicts the opsonic activity of the chimeric monoclonal antibody 96-
  • Figure 18 demonstrates the enhancement of survival after administration of
  • MAB 96-110 in a lethal model of S. epidermidis sepsis.
  • Figure 19 depicts the effect of the chimeric monoclonal antibody 96-110 on the survival of adult mice after intraperitoneal challenge with S. epidermidis.
  • Figure 20 sets forth the effect of the chimeric monoclonal antibody 96-110 on
  • Figure 21 depicts bacteremia levels four hours after infection with S. epidermidis at different doses of the chimeric monoclonal antibody 96-110.
  • Figure 22 sets forth the effect of the chimeric monoclonal antibody 96-110 on
  • the present invention provides anti-lipoteichoic acid (LTA) murine antibodies (including monoclonal antibodies) and chimeric murine-human antibodies, and
  • fragments, derivatives, and regions thereof which bind to and opsonize whole Gram positive cocci such as Staphylococcus to thereby enhance phagocytosis and killing of such bacteria in vitro and which enhance protection from lethal infection of such bacteria in vivo.
  • invention preferably recognize and bind to an epitope of LTA that can block the
  • epithelial cells such as human epithelial cells.
  • the invention provides broadly reactive and opsonic antibodies for the diagnosis, prevention, and/or treatment of bacterial infections caused by Gram
  • the antibodies of the invention are broadly reactive with Gram positive bacteria, meaning that they selectively recognize and bind to Gram positive bacteria and do not recognize or bind to Gram negative bacteria. Any conventional binding assay can be used to assess this binding, including for example, the enzyme linked
  • Teichoic acids are polymers of either giycerol phosphate or ribitol phosphate with various sugars,
  • teichoic acids are lipoteichoic acids which are
  • teichoic acids made up of giycerol phosphate which is primarily linked to a glycolipid in the underlying cell membrane.
  • lipoteichoic acids that are characteristically surface exposed on Gram positive bacteria.
  • the antibodies of the invention are also opsonic, or exhibit opsonic activity,
  • an opsonin generally either an antibody or the serum factor C3b
  • Opsonic activity may be measured in any conventional manner as described below.
  • anti-LTA antibodies of the invention The ability of the anti-LTA antibodies of the invention to bind to and opsonize Gram positive bacteria and thereby enhance phagocytosis and cell killing in vitro and
  • anti-LTA antibodies have been reported to lack opsonic activity. Indeed, anti-LTA antibodies have been often used as controls.
  • S. epidermidis has become a major cause of nosocomial infections in patients whose treatments include the placement of foreign objects; S. epidermidis has become a common cause of peritonitis in patients with continuous ambulatory peritoneal dialysis; and S. epidermidis is now recognized as a common
  • opsonic antibody which specifically reacts in an assay with S. epidermidis serotypes I, II and III, and which exhibits opsonic activity greater than 70%.
  • the In Vitro Methods describes the use of a Serotype II S. epidermidis, such as the Hay strain, that identifies pathogenic Staphylococcus infections.
  • mice were immunized with whole strain Hay S. epidermidis from which hybridomas were produced.
  • the antibodies of one clone first designated 96-105CE11 IF6 and later designated 96-110 MAB
  • 96-105CE11 IF6 a strong IgG reaction
  • 96-110 MAB a strong IgG reaction
  • strain Hay to all three serotypes of S. epidermidis, to S. hemolyticus, S. hominus, and two serotypes of S. aureus (Tables 3-6) but not to the Gram negative control, Haemophiius influenza.
  • the antibody of the present invention exhibits very strong binding, i.e., O.D.s of around twice background in an enzyme-linked immunosorbent assay (described below), against strain Hay.
  • the level of high binding is equal to or greater than five times background. In other embodiments, the
  • level of high binding is equal to or greater than 10 times background.
  • background the level observed when all the reagents other than the antibody being tested
  • enhanced refers to activity that measurably exceeds background at a valuable level.
  • the level deemed valuable may well vary depending on the specific circumstances of the infection, including the type of bacteria and the severity of the infection. For example, for enhanced opsonic or phagocytic activity, in a
  • an enhanced response is equal to or greater than 75% over background. In another preferred embodiment, the enhanced response is equal to or greater than 80% over background. In yet another embodiment, the enhanced response is equal to or greater than 90% over background.
  • MAB 96-110 was assessed in a lethal infection model in both neonatal rats and adult mice. As set forth in Example 3, survival in control animals given either no therapy, saline, or control MAB, ranged
  • MAB 96-110 enhanced the survival to 50% or
  • the enhancement measured is of survival
  • the preferred increase over background may be more modest than above.
  • survival of 25% may be an enhanced response.
  • survival may be greater than 50%. Again, the person of skill in the art would recognize other meaningful increases in survival as within the invention.
  • an "antigen” is a molecule or a portion of a molecule capable of being bound by an antibody and which is also capable of inducing an animal to produce antibody capable of binding to an epitope of that antigen.
  • An antigen may have one or more epitopes.
  • An “epitope” analogously means that portion of the molecule that is capable of being recognized by and bound by an antibody. In general, epitopes consist of chemically active surface groupings of molecules such
  • amino acids or sugar side chains that have specified three dimensional structural and specific charge characteristics.
  • one aspect of the present invention involves a peptide having the sequence
  • the epitope of the invention may be identical to one of these sequences or may be substantially homologous to these sequences such that the anti-LTA antibodies of the invention will bind to them.
  • the substantially homologous sequences of the invention are those that are able to induce the anti-
  • LTA antibodies of the invention LTA antibodies of the invention.
  • Other peptide epitope mimics within the invention may vary in length and sequence from these two peptides.
  • the present invention also encompasses recombinant epitopes, epitope
  • antigen can be identified, isolated, cloned, and transferred to a prokaryotic or
  • the present invention includes antibodies that are capable of binding to the LTA of Gram positive bacteria, including both coagulase negative and coagulase positive bacteria, and of enhancing the opsonization of such bacteria.
  • LTA antibodies include polyclonal antibodies as well as monoclonal antibodies
  • hybridomas of the invention such as MAB 96-110 as well as other
  • the strength of the binding may range from twice above background, to five- and ten-times above background.
  • the antibodies, fragments, regions, and derivatives of the present invention are capable of enhancing the opsonization of such bacteria, at rates ranging from 75% and up.
  • fragments of the antibodies of the invention include, for example, Fab,
  • Fab', F(ab') 2 , and SFv fragments are produced from intact antibodies using methods well known in the art such as, for example, proteolytic cleavage with enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab') 2 ).
  • proteolytic cleavage with enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab') 2 ).
  • the "regions" of the antibodies of the present invention include a heavy chain
  • the light chains may be either a lambda or a kappa chain.
  • the regions include at least one heavy
  • variable region or a light chain variable region which binds a portion of LTA, including for example the specific antigen binding sites (that which binds to the epitope) of the two regions.
  • these two variable regions can be linked together as a single chain antibody. While a full length heavy chain may
  • the antibody fragments encompassing the variable regions may be suitable for inhibition of bacterial binding to epithelial cells and may also be anti-inflammatory.
  • the antibody is a chimeric
  • mouse/human antibody made up of regions from the anti-LTA antibodies of the
  • This humanized or chimeric heavy chain may be combined with a chimeric L chain that comprises the antigen binding region of the light chain variable region of the anti-LTA antibody linked to at least a portion of the human light
  • chimeric antibodies of the invention may be monovalent, divalent, or polyvalent immunoglobulins.
  • a monovalent chimeric antibody is a dimer (HL) formed by a chimeric H chain associated through disulfide bridges with a
  • a divalent chimeric antibody is a tetramer (H 2 L 2 ) formed by two HL dimers associated through at least one disulfide bridge.
  • a polyvalent chimeric antibody is based on an aggregation of chains.
  • Examples 8-10 which set forth in detail the preparation of a preferred chimeric IgG antibody (and in Examples 11-13 which describe the functional activity of this preferred chimeric anibody).
  • the heavy chain constant region can be an IgM or IgA antibody.
  • inventions also encompasses the DNA sequence of the gene coding for the antibodies as well as the peptides encoded by the DNA. Particularly preferred DNA and peptide sequences are set forth in Figure 12. That figure provides the variable
  • CDR Complementarity Determining Regions
  • the invention includes these DNA and peptide sequences as well as DNA and peptide sequences that are homologous to these sequences. In a preferred embodiment, these sequences are 70 % homologous although other
  • preferred embodiments include sequences that are 75%, 80%, 85%, 90%, and 95%
  • DNA sequences of the invention can be identified, isolated, cloned, and
  • the CDR can be graphed onto any human antibody frame using techniques standard in the art, in such a manner that the CDR maintains the same binding specificity as in
  • DNA and peptide sequences of the antibodies of the invention may form the basis of
  • antibody “derivatives” which include, for example, the proteins or peptides encoded by truncated or modified genes. Such proteins or peptides may function similarly to the antibodies of the invention. Other modifications, such as the addition of other sequences that may enhance the effector function, which includes phagocytosis
  • the present invention also discloses a pharmaceutical composition
  • a pharmaceutical composition comprising the anti-LTA antibodies, whether polyclonal, monoclonal or chimeric, as
  • compositions of the invention may alter- natively comprise the isolated antigen, epitope, or portions thereof, together with a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers can be sterile liquids, such as water, oils, including petroleum oil, animal oil, vegetable oil, peanut oil, soybean oil, mineral oil, sesame oil, and the like. With intravenous administration, water is a preferred carrier. Saline solutions, aqueous dextrose, and giycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, 18th Edition (A. Gennaro, ed., Mack Pub., Easton, Pa., 1990), incorporated by ref ⁇
  • the present invention provides methods for treating a patient infected with, or suspected of being infected with, a Gram positive bacteria such as a staphylococcal organism.
  • the method comprises administering a therapeutically effective amount of a pharmaceutical composition comprising the anti-LTA immunoglobulin (whether polyclonal or monoclonal or chimeric, including fragments, regions, and derivatives thereof) and a pharmaceutically acceptable carrier.
  • a patient can be a human or other mammal, such as a dog, cat, cow, sheep, pig, or goat.
  • the patient is preferably a human.
  • therapy as above or as described below may be primary or supplemental to additional treatment, such as antibiotic therapy, for a staphylococcal infection, an
  • a further embodiment of the present invention is a method of preventing such
  • infections comprising administering a prophyiactically effective amount of a pharmaceutical composition comprising the anti-LTA antibody (whether polyclonal or monoclonal or chimeric, including fragments, regions, and derivatives thereof) and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising the anti-LTA antibody (whether polyclonal or monoclonal or chimeric, including fragments, regions, and derivatives thereof) and a pharmaceutically acceptable carrier.
  • Such - therapy as above or as described below may be primary or supplemental to additional treatment, such as antibiotic therapy, for a staphylococcal infection, an infection caused by a different agent, or an unrelated disease. Indeed, combination therapy with other antibodies is expressly contemplated within the invention.
  • the peptide which mimics the LTA epitope would be useful to prevent binding of Gram positive bacteria to epithelial cells and thereby inhibit colonization.
  • peptide may be administered intranasally to prevent an infection or to minimize a current infection.
  • Yet another preferred embodiment of the present invention is a vaccine comprising the epitope, epitope mimic, or other part of the LTA antigen and a pharmaceutically acceptable carrier. Upon introduction into a host, the vaccine
  • the vaccine may include the epitope, an epitope mimic, any
  • Vaccinations are particularly beneficial for individuals known to be or suspected of being at risk of infection by Gram positive bacteria. This includes patients receiving body implants, such as valves, patients with indwelling catheters, patients preparing to undergo surgery involving breakage or damage of skin or mucosal tissue, certain health care workers, and patients expected to develop
  • Treatment comprises administering the pharmaceutical composition (including antibodies and vaccines) by intravenous, intraperitoneal, intracorporeal injection, intra-articular, intraventricular, intrathecal, intramuscular, subcutaneous, intranasally, intravaginally, orally, or by any other effective method of administration.
  • the composition may also be given locally, such as by injection to the particular area infected, either intramuscularly or subcutaneously.
  • Administration can comprise
  • the treatment can also be applied to objects to be placed within a patient, such as dwelling catheters, cardiac values, cerebrospinal fluid shunts, joint prostheses, other implants into the body, or any other objects, instruments, or appliances at risk of becoming infected with a Gram positive
  • compositions of the invention including all anti-LTA antibodies (whether polyclonal or monoclonal or
  • LTA antigen and vaccines based on such epitope or antigens are the reduction in cytokine release that results from the introduction of the LTA of a Gram positive bacteria.
  • LTA induces cytokines, including for example tumor necrosis factor alpha, Interleukin 6, and interferon gamma. See Takada et al., Infection and Immunity, 63 (1):57-65 (January 1995). Accordingly, the
  • compositions of the invention may enhance protection at three levels: (1) by binding to LTA on the bacteria and thereby blocking the initial binding to epithelial cells and preventing subsequent invasion of the bacteria; (2) by enhancing opsonization of the bacteria and thereby enhancing clearance of the bacteria from tissues and blood;
  • Hybridoma 96-110 was deposited at the ATCC on June 13, 1997 under Accession No. HB-12368.
  • Isotype was determined using an isotype kit obtained from Zymed Laboratories. The kit can be ordered under number 90-6550.
  • immunoglobulin is reacted with a
  • the binding assay is preferably an enzyme-linked immunosorbent assay (ELISA) or a radioimmunoassay (RIA), but may also be an agglutination assay, a coagglutination assay, a colorimetric assay, a fluorescent binding assay, or any other suitable binding assay that is known in the
  • the assay can be performed by competitive or noncompetitive procedures with results determined directly or indirectly.
  • the Staphylococcus preparation may be fixed to a suitable solid support
  • the solid support is preferably a titration plate.
  • the fixed preparation is incubated
  • immunoglobulin which is isolated or in a biological fluid such as ascites, and
  • a positive reaction occurs when the amount of binding observed for the test sample is greater than the amount of binding for a negative control.
  • a negative control is any sample known not to contain antigen-specific immunoglobulin. Positive binding may be determined from a simple
  • This series may include samples containing measured amounts of immunoglobulin that specifically bind to the fixed antigen, creating a standard curve from which the amount of antigen-specific immunoglobulin in an unknown sample can be determined.
  • antibody can be fixed to a solid support and im ⁇
  • munoglobulin identified by its ability to bind a bacterial preparation bound to the fixed antibodies.
  • Supematants were tested at a dilution of 1 :2. Ascites and purified antibody were tested at dilutions indicated in the Tables. After addition of the antibody, the wells were incubated at room temperature for 30-60 minutes in a draft-free environment.
  • detection antibody rabbit anti-mouse IgG, conjugated to horse radish peroxidase (HRP) and diluted 1 :4000 in PBS-T.
  • the detection antibodies were specific for mouse gamma, mu or alpha heavy chains (Zymed catalog numbers 61-6020, 61- 6820 or 61-6720 respectively), as indicated in the Tables.
  • TMB substrate solution 100 ul of one-component TMB substrate solution (Kirkegaard and Perry Labs catalog number 50-76-05).
  • the wells were incubated in the dark at room temperature for 15 minutes.
  • the reaction was stopped by the addition of 80 ul of TMB stop solution (Kirkegaard and Perry Labs catalog number 50-85-05) and the absorbance of each well at 450 nm was determined using a Molecular Devices Vmax plate reader.
  • LTA's were obtained from Sigma Chemical Company and diluted in PBS to 1 ug/ml. One hundred microliters of the 1 ug/ml solution was distributed into replicate Nunc Maxisorp Stripwells. The LTA was incubated in the wells overnight at room temperature. The unbound material was
  • the immunoassay wells were coated with bacteria as described above.
  • the purified monoclonal antibody (MAb) was reacted with a solution of recombinant Protein A conjugated to HRP and diluted 1 :500 in PBS-T. This reaction was allowed to proceed for 30 minutes.
  • the Protein A-HRP-MAb was allowed to react in the coated wells for 30-60 minutes at room temperature. The wells were then washed with PBS-T and TMB substrate solution was added and the assay completed as described above.
  • An opsonization assay can be a colorimetric assay, a chemiluminescent assay, a fluorescent or radiolabel uptake assay, a cell-mediated bactericidal assay, or any other appropriate assay known in the art which measures the opsonic
  • an infectious agent a substance that identifies broadly reactive immunoglobulin.
  • an infectious agent a substance that identifies broadly reactive immunoglobulin.
  • the opsonizing substance to be tested or an opsonizing substance plus a purported opsonizing enhancing substance.
  • the opsonizing substance to be tested or an opsonizing substance plus a purported opsonizing enhancing substance.
  • opsonization assay is a cell-mediated bactericidal assay.
  • the in vitro assay the in vitro assay
  • an infectious agent typically a bacterium
  • a bacterium typically a bacterium
  • phagocytic cell and an opsonizing substance, such as immunoglobulin.
  • an eukaryotic cell with phagocytic or binding ability may be used in a cell-mediated bactericidal assay, a macrophage, a monocyte, a neutrophil, or any combination of these cells, is preferred.
  • Complement proteins may be included to promote the production of phagocytic cells.
  • the opsonic ability of immunoglobulin is determined from the amount or number of infectious agents remaining after incubation. In a cell-mediated bactericidal assay, this is accomplished by comparing the number of surviving bacteria between two similar assays, only one of which contains the purported
  • the opsonic ability is determined by measuring the numbers of viable organisms before and after incubation. A reduced number of bacteria after incubation in the presence of immunoglobulin indicates a positive opsonizing ability. In the cell-mediated bactericidal assay, positive
  • opsonization is determined by culturing the incubation mixture under appropriate bacterial growth conditions. Any significant reduction in the number of viable bacteria comparing pre- and post-incubation samples, or between samples which contain immunoglobulin and those that do not, is a positive reaction.
  • Another preferred method of identifying agents for the treatment or prevention of a infection by Gram positive bacteria employs lethal models of sepsis that measure clearance and protection.
  • agents can be immunoglobulin or other
  • a particularly useful animal model comprises administering an antibody and a
  • Gram positive organism to an immunocompromised (e.g., an immature) animal, followed by evaluating whether the antibody reduces mortality of the animal or enhances clearance of the organism from the animal.
  • This assay may use any immature animal, including the rabbit, the guinea pig, the mouse, the rat, or any other suitable laboratory animal.
  • the suckling rat lethal animal model is most preferred.
  • Such a model can readily incorporate an infected foreign body, such as an infected catheter, to more closely mimic the clinical setting.
  • An alternative model utilizes adult susceptible animals, such as CF1 mice.
  • Clearance is evaluated by determining whether the pharmaceutical composition enhances clearance of the infectious agent from the animal. This is
  • results are considered positive if the pharmaceutical composition enhances
  • mice were immunized with whole S. epidermidis, Strain Hay.
  • a suspension of heat killed S. epidermidis was adjusted to an optical density
  • mice obtained from Harlan Sprague Dawley (Indianapolis, IN) were immunized subcutaneously with 0.2 mis of the immunogen described above. The mice received a booster immunization approximately two and
  • mice 8159 was removed and used for hybridoma formation.
  • Hybridomas were prepared by the general methods of Shulman, Wilde and Kohler Nature 276:269-270 (1978) and Bartal and Hirshaut "Current Methods in
  • Hybridoma Formation in Methods of Hybridoma Formation Bartal and Heishaut, eds., Humana Press, Clifton, New Jersey (1987). A total of 2.135 X 10 8 spleenocytes from mouse 8159 were mixed with 2.35 X 10 7 SP2/0 mouse myeloma
  • the hybridoma cell suspension was gently resuspended into the medium and the cells pelleted by centrifugation (500 X g, 10 minutes at room temperature). The supernatant was removed and the cells resuspended in medium RPMI 1640, supplemented with 10% heat-inactivated fetal bovine serum, 0.05 mM hypoxanthine and 16 uM thymidine (HT medium).
  • medium RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum, 0.05 mM hypoxanthine and 16 uM thymidine (HT medium).
  • One hundred ul of the hybridoma cells were planted into 760 wells of 96-well tissue culture plates. Eight wells (column 1 of plate A) received approximately 2.5 X 10 4 SP2/0 cells in 100 ul.
  • the SP2/0 cells were planted into 760 wells of 96-well tissue culture plates. Eight wells (column 1 of plate A) received approximately 2.5 X 10
  • peroxidase labeled protein A was mixed with the purified 96-110 MAB and then reacted with S. aureus type 5 (SA5) and S. aureus type 8 (SA8) obtained from ATCC at Accession Nos. 12602 and 12605, respectively. Both S. aureus serotypes reacted strongly with the 96-110
  • Antibodies which bind to an antigen may not necessarily enhance opsonization or enhance protection from infection. Therefore, a neutrophil mediated
  • bactericidal assay was used to determine the functional activity of antibody to S. epidermidis.
  • Neutrophils were isolated from adult venous blood by dextran sedimentation and ficoll-hypaque density centrifugation. Washed neutrophils were added to round-bottomed wells of microtiter plates (approximately 10 6 cells per well) with approximately 3 x 10 4 mid-log phase bacteria (S. epidermidis Hay, ATCC 55133). Newborn lamb serum (10 uls), screened to assure absence of antibody to S. epidermidis, was used as a source of active complement.
  • Staphylococcus (S. epidermidis) to 100%, compared with 49.5% with C and PMN alone (Table 7).
  • the coagulase positive Staphylococcus also showed enhanced phagocytosis at 1 :10 and 1 :40 dilution (83.3% and 78.9% respectively) compared
  • Such an antibody would be capable of promoting clearance of Staphylococci that have invaded a host and would be useful therapeutic agent.
  • Antibody Purified M X Hay, 96-110
  • Opsonic antibody correlates with enhanced protection from staphylococcal infections, as set forth in the recent series of Fischer applications and issued patent described and incorporated by reference above.
  • studies were conducted using lethal infection models.
  • MAB 96-110 enhanced survival in this lethal neonatal model of coagulase positive staphylococcus sepsis (Figure 3): 8/15 survived after treatment with MAB 96-110, and 0/10 survived with Control MAB or 2/25 with saline treatment.
  • CF1 mice were given 0.5 ml S. epidermidis (Hay) IP (3.5 x 10 9 bacteria). About 24 hrs and 2 hrs before and 24 hrs post- infection, 320 ug of MAB 96-110 were given to one group of mice and compared with a second group infected in the same manner, but not treated with antibody. All
  • MAB 96-110 could enhance survival in lethal coagulase positive and
  • the original libraries were acquired from George P. Smith, Division of Biological Sciences, University of Missouri, Columbia, MO.
  • the 96-110 antibody was crosslinked to Biotin using the Sulfo-NHS-biotin ester crossiinking kit following the
  • TBS/Tween 50mM Tris-HCI pH 7.5, 150mM NaCl, 0.5% v/v Tween 20. Incubate dishes overnight at 4°C rocking with 400ul TBS/Tween
  • the second and third round of panning are carried out the same way.
  • the input and resultant phage are titered to determine yield from biopanning.
  • the titered infection plates are used to pick
  • NZY medium containing 40ug/ml tetracycline. Replicative form DNA is extracted
  • the MAB 96-110 was also analyzed for binding to LTA from S. pyogenes
  • group A streptococcus group A streptococcus
  • various group A streptococcal M types The MAB showed strong binding to the LTA and also bound to the different M types with strongest binding to M1 and M3 (Table 10).
  • LTA induces proinflammatory cytokines such as TNF, IL-6 and Interferon gamma
  • MABs with strong anti-LTA binding will also have an anti-inflammatory action and modulate cytokine production secondary to LTA bearing bacteria.
  • Anti-LTA antibodies or vaccines could be designed and produced to modulate cytokine production and inflammation in tissues and prevent the
  • Substrate was TMB.
  • the hybridoma cell producing the 96-110 antibody was obtained as described above. A vial of cells was thawed, washed with serum free medium and then resuspended in IMDM (Mediatech) complete media supplemented with 10%FBS
  • RNA was isolated from 1 10 8 96-110 cells using the Midi RNA Isolation kit (Qiagen) following the manufacturer's procedure. The RNA was dissolved in 10mM Tris, 0.1 mM EDTA (pH8.4) containing 0.03U/ ⁇ g Prime RNase Inhibitor (5'-3') to a final concentration of 0.25 ⁇ g/ ⁇ l.
  • Figure 10 shows the strategy for cloning the variable region gene fragments and Figure 11 lists the oligonucleotides primers used.
  • the 96-110 total RNA (2 ⁇ g) was converted to cDNA by using Superscript ll-MMLV Reverse transcriptase (Life Technologies) and mouse kappa (OKA57) and mouse heavy chain (JS160-162)- specific priming according of manufacturer's procedures.
  • the first strand cDNA synthesis products were then purified using a Centricon-30 concentrator device (Amicon). Of the 40 ⁇ l of cDNA recovered, 5 ⁇ l was used as template DNA for PCR.
  • Typical PCR amplification reactions (100 ⁇ l) contained template DNA, 50 pmoles of
  • reaction buffer 200 ⁇ M dNTP, 1mM MgCI 2 .
  • the template was denatured by an initial incubation at 96°C for 5 min.
  • the products were amplified by 15 thermal
  • the heavy chain PCR products (approximately 400 bp) were then cloned into a bacterial vector for DNA sequence determination. Ligations of the PCR fragments were carried out into the PCR2.1 (invitrogen) T/A style cloning vector following the
  • the light chain PCR products were treated differently.
  • the hybridoma cell line that expresses the 96-110 antibody was made by fusing mouse spleenocytes with
  • the SP20 myeloma cell line transcribes a pseudogene for the kappa light chain.
  • the hybridoma cell line that expresses the 96-110 antibody transcribes a second pseudogene product for a kappa light chain that apparently arose from the spleenocyte partner of the hybridoma fusion event.
  • second pseudogene transcript can be expressed from an expression vector
  • pseudogene transcripts when converted to cDNA by RT-PCR, contain an Af/lll
  • variable region was digested with Af/lll and those products that did not cut were then cloned into the pGEM T-Easy (Promega) T/A style cloning vector using the
  • Light chain candidate (pJRS319) clones were digested with EcoRI (New England Biolabs) using the manufacturer's procedures to identify clones containing inserts of the appropriate size (350bp).
  • EcoRI New England Biolabs
  • the final consensus DNA sequence of the light chain variable regions is shown in Figure 12.
  • the amino acids encoded by these sequences match the N-terminal amino acid analyses of the heavy and light chain peptide fragments produced by the hybridoma cell line.
  • the heavy and light chain variable regions were then subcloned into a mammalian expression plasmid vector pSUN 15 for production of recombinant chimeric antibody molecules.
  • the creation of the expression vector was an extensive process of DNA fragment ligations and site directed mutagenesis steps.
  • the result was a vector that expresses both antibody chains with CMV promoter driven transcription.
  • Neomycin resistance serves as a dominant selectable marker for transfection of mammalian cells.
  • it has been designed to allow convenient cloning of any light chain variable region as EcoRVISs BI fragment, any heavy chain variable region as a Nrul/EcoRI fragment, and any heavy chain constant domain as an EcoRIINotl fragment. These restriction sites were chosen because
  • the backbone of the vector was the plasmid pCDNA3 (Invitrogen). This
  • a second "heavy chain vector" was constructed from the pCDNA3mut.
  • LCPL.LCVK plasmid by replacing the light chain expression region (Hindlll/Xhol) with a "heavy chain polylinker" consisting of restriction sites Hpal, BspEI, EcoRV, Kpnl, and Xhol.
  • a Smal/ Kpnl DNA fragment contains a heavy chain leader, antiCKMB lgG2b chain genomic fragment.
  • a Kpnl/Sall oligo nucleotide fragment containing a 3' UTR and a NotI upstream of the Sail site was subsequently cloned into the KpnUXhol digested plasmid, (knocking out the Xhol site) to create the plasmid pCDNA3mut.HCPLHCV2b.
  • CKMB variable or constant domain DNA sequences This was done by cutting the plasmid pCDNA3mut.LCPL.LCVK with EcoRV/Xhol and inserting a linker oligonucleotide fragment containing EcoRV, BstBI, and Xhol sites to create pSUN9.
  • a human kappa light chain constant domain was then cloned into pSUN9 as a BstB/Xhol fragment, and a human lgG1 constant domain was cloned into pSUNIO as a EcoRI/NotI fragment.
  • a Bglll/Nhel fragment from the human heavy chain vector was then cloned into the human light chain vector cut with BamHI/Nhel to create pSUNI ⁇ .
  • This vector results in the production of recombinant antibody molecules under
  • the heavy chain molecules are direct cDNA constructs that fuse the variable region sequence directly into the human lgG1 constant domain.
  • the light chain molecules on the other hand, have a mouse kappa intron region 3' of the variable region coding fragment. After splicing the variable region becomes fused to a human kappa constant region exon.
  • the selectable marker for the vector in mammalian coils is Neomycin (G418).
  • variable region gene fragments were re-amplified by PCR using primers that adapted the fragments for cloning into the expression vector (see Figures 12 and 14).
  • the heavy chain front primer (96110HF2) includes a 5' tail that encodes the C-terminus of the heavy chain leader and an Nrul restriction site for cloning, while the heavy chain reverse primer (96110HB) adds a 3' EcoRI restriction site for cloning.
  • the light chain front primer (96110bLF) converts the first amino acid of the heavy chain leader and an Nrul restriction site for cloning.
  • the light chain reverse primer (96-110bLB) adds a 3' DNA sequence for the joining region-kappa exon splice junction followed by a
  • the reverse primer introduces a
  • PCRs were performed as described above except 10ng of plasmid template was used in each case. Following a 5 min. incubation at 96 °C, the PCR perimeters were 35 thermal cycles of 58°C for 30 sec, 70°C for 30 se , and 96°C for 1 min.
  • the 96-110 heavy chain PCR product (approximately 400 bp) was digested with Nrul and EcoRI (New England Biolabs), purified using a Qiaquick PCR Purification column (Qiagen), as described by the manufacturer, and ligated into Nrul I EcoRI digested and gel-purified pSUN15, resulting in plasmid pJRS311 (see Figure
  • Antibody production assays were preformed in 8-well strips from 96-well microtiter plates (Maxisorp F8; Nunc, Inc.) coated at a 1 :500 dilution with Goat anti- Human IgG antibody (Pierce) using a bicarbonate coating buffer, pH 8.5. The plates are covered with pressure sensitive film (Falcon, Becton Dickinson) and incubated
  • the activity assays were preformed in 8-well strips from 96-well microtiter plates (Maxisorp F8; Nunc, Inc.) coated at 0.09 OD/well with heat-killed Staph Hay cell material suspended in MeOH. The plates are left uncovered and incubated
  • sample/conjugate diluent 100 microliters was added to the samples, then incubated
  • the plasmid pJRS334-1 was transfected into NS/0 cells (obtainable from Baxter International) and CHO cells using electroporation.
  • the plasmid was linearized with Pwvl restriction digestion. 25 micrograms of digested plasmid DNA
  • Neutrophils specifically polymorphonuclear neutrophils, were isolated from adult venous blood by dextran sedimentation and ficoll-hypaque density centrifugation. Washed neutrophils were added to round-bottomed wells of microtiter plates (approximately 10 6 cells per well) with approximately 3 x 10 4 mid-log phase bacteria (S. epidermidis Hay, ATCC 55133). Human sera (10 uls), screened to assure absence of antibody to S. epidermidis, was used as a source of active complement (C-Barb-Ex (1:4)).
  • Bacteremia is expressed as the number of bacteria isolated on blood agar after a 1 : 1000 dilutions. As set forth in Figure 20, the chimeric MAB reduced bacterial levels by over 2 logs. Additional studies demonstrated that bacteremia was reduced to a greater degree using 40 mg/kg/dose compared to 20 mg/kg/dose even if survival was comparable. See Figure 21.
  • Wistar rats were treated with lipid emulsion (as is standard in newborn care for nutritional purposes) 0.2 ml, 20% IP on day -1 and again on day +1 and +2 to induce further compromise of the immuno system.
  • lipid emulsion as is standard in newborn care for nutritional purposes
  • Table 11 SQ through a plastic catheter and the catheter was left in place under the skin. Saline, 0.2 ml, or MAB 96-110, 0.2 ml (dose of 50-60 mg/kg), was given IP 30 min before and 24 hours after infection. The animals were followed for 5 days.
  • antibody to LTA provides prophylactic and therapeutic capabilities against staphylococcal infections and vaccines using LTA or peptide mimeotopes of LTA that induce anti- LTA antibodies would also have prophylactic capabilities.
  • ADDRESSEE FINNEGAN, HENDERSON, FARABOW, GARRETT &
  • Trp His Trp Arg His Arg lie Pro Leu Gin Leu Ala Ala Gly Arg 1 5 10 15
  • MOLECULE TYPE other nucleic acid
  • GGG ANT CAT GCG GAT AGG GTT TAT GGG GCC 30
  • Gly Ala Trp His Trp Arg His Arg lie Pro Leu Gin Leu Ala Ala Gly 1 5 10 15

Abstract

The present invention encompasses monoclonal and chimeric antibodies that bind to lipoteichoic acid of Gram positive bacteria. The antibodies also bind to whole bacteria and enhance phagocytosis and killing of the bacteria in vitro and enhance protection from lethal infection in vivo. The mouse monoclonal antibody has been humanized and the resulting chimeric antibody provides a previously unknown means to diagnose, prevent and/or treat infections caused by gram positive bacteria bearing lipoteichoic acid. This invention also encompasses a peptide mimic of the lipoteichoic acid epitope binding site defined by the monoclonal antibody. This epitope or epitope peptide mimic identifies other antibodies that may bind to the lipoteichoic acid epitope. Moreover, the epitope or epitope peptide mimic provides a valuable substrate for the generation of vaccines or other therapeutics.

Description

OPSONIC AND PROTECTIVE MONOCLONAL AND CHIMERIC ANTIBODIES SPECIFIC FOR LIPOTEICHOIC ACID OF GRAM POSITIVE BACTERIA
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of Provisional Patent Application Serial No. 60/049,871 , filed June 16, 1997, which application is specifically incorporated herein by reference.
FIELD OF THE INVENTION
This invention in the fields of immunology and infectious diseases relates to
antibodies that are specific for Gram positive bacteria, particularly to lipoteichoic acids exposed on the surface of the bacteria. The invention includes monoclonal and chimeric antibodies, as well as fragments, regions and derivatives thereof. This
invention also relates to the epitope to which the antibodies of the invention bind as well as the sequences, fragments, and regions of the epitopes. Both the antibodies and peptides that encompass the epitope, and regions and fragments thereof, may be used for diagnostic, prophylactic and therapeutic applications.
BACKGROUND OF THE INVENTION
Man has long battled bacterial infections, and no one can doubt the tremendous successes obtained. Before the discovery and development of
antibiotics, death due to a bacterial infection was frequently rapid and inevitable.
Surgical procedures and sanitary conditions have vastly improved from the time
when amputation was associated with a 50 percent mortality rate.
Nonetheless, the battle has not been won. Undoubtedly a significant part of
the problem is that bacteria are the product of nearly 3 billion years of natural
selection from which they have emerged as an immensely diverse group of organisms that colonize almost all parts of the world and its inhabitants. To begin to
understand bacteria requires categorization, and the most fundamental categories for bacteria are their response to the Gram stain, yielding (for the most part) Gram positive bacteria and Gram negative bacteria.
The difference in response to the Gram stain results from differences in
bacterial cell walls. The cells walls of Gram negative bacteria are made up of a unique outer membrane of two opposing phospholipid-protein leaflets, with an ordinary phospholipid in the inner leaflet but the extremely toxic lipopolysaccharide in the outer leaflet. The cell walls of Gram positive bacteria seem much simpler in
comparison, containing two major components, peptidoglycan and teichoic acids plus additional carbohydrates and proteins depending on the species.
Of the Gram positive bacteria, one of the most common genera is Staphylococcus. Staphylococci commonly colonize humans and animals and are an
important cause of human morbidity and mortality, particularly in hospitalized patients. Staphylococci are prevalent on the skin and mucosal linings and, accordingly, are ideally situated to produce both localized and systemic infections.
There are two main groups of Staphylococci divided according to the
production of "coagulase," an enzyme that causes fibrin to coagulate and to form a
clot: coagulase positive and coagulase negative. The coagulase positive Staphylococcus species most frequently pathogenic in humans is Staphylococcus aureus. S. aureus is the most virulent Staphylococcus and produces severe and
often fatal disease in both normal and immunocompromised hosts. Staphylococcus
epidermidis is the most common coagulase negative species. In recent years, S. epidermidis has become a major cause of nosocomial infection in patients whose treatments include the placement of foreign objects such as cerebrospinal fluid shunts, cardiac valves, vascular catheters, joint prostheses, and other implants into the body. S. epidermidis and S. aureus are common causes of post-operative wound infections and S. epidermidis has also become a common
cause of peritonitis in patients with continuous ambulatory peritoneal dialysis. In a similar manner, patients with impaired immunity and those receiving parenteral nutrition through central venous catheters are at high risk for developing S.
epidermidis sepsis. (C.C. Patrick, J. Pediatr., 116:497 (1990)). S. epidermidis is
now recognized as a common cause of neonatal nosocomial sepsis. Infections frequently occur in premature infants that have received parenteral nutrition which can be a direct or indirect source of contamination.
Staphylococcal infections are difficult to treat for a variety of reasons.
Resistance to antibiotics is common and becoming more so. See L. Garrett, The Coming Plaque. "The Revenge of the Germs or Just Keep Inventing New Drugs" Ch. 13, pgs. 411-456, Farrar, Straus and Giroux, NY, Eds. (1994). In one study, the
majority of Staphylococci isolated from blood cultures of septic infants were multiply
resistant to antibiotics (A. Fleer et al., Pediatr. Infect. Dis. 2:426 (1983)). A more recent study describes methiciilin-resistant S. aureus (J. Romero-Vivas, et al., Clin. Infect. Dis. 21 :1417-23 (1995)) and a recent review notes that the emergence of
antibiotic resistance among clinical isolates makes treatment difficult (J. Lee.,
Trends in Micro. 4(4): 162-66 (April 1996). Recent reports in the popular press also
describe troubling incidents of antibiotic resistance. See The Washington Post "Microbe in Hospital Infections Show Resistance to Antibiotics," May 29, 1997; The
Washington Times, "Deadly bacteria outwits antibiotics," May 29, 1997.
In addition, host resistance to Staphylococcal infections is not clearly
understood. Opsonic antibodies have been proposed to prevent or treat
Staphylococcal infections. See U.S. Patent No. 5,571 ,511 to G.W. Fischer issued
November 5, 1996, specifically incorporated by reference. The microbial targets for
these antibodies have been capsuiar polysacchahdes or surface proteins. As to
capsular polysaccharides, the immunization studies of Fattom et al., J. Clin. Micro.
30(12):3270-3273 (1992) demonstrated that opsonization was related to S.
epidermidis type-specific anti-capsular antibody, suggesting that S. epidermidis and
S. aureas have a similar pathogenesis and opsonic requirement as other
encapsulated Gram positive cocci such as Streptococcus pneumonia. As to surface
proteins, Timmerman, et al., J. Med. Micro. 35:65-71 (1991) identified a surface
protein of S. epidermidis that was opsonic for the homologous strain used for
immunization and for monoclonal antibody production. While other monoclonal
antibodies were identified that bound to non-homologous S. epidermidis strains, only
the monoclonal antibody produced to the homologous strain was opsonic and
opsonization was enhanced only to the homologous strain but not to heterologous
strains. Accordingly, based on the studies of Fattom et al., and Timmerman et al.,
and others in the field (and in contrast to our own studies), one would not expect that
an antibody that is broadly reactive to multiple strains of S. epidermidis and to S.
aureus would have opsonic activity against both. This is particularly true for
antibodies that bind to both coagulase positive and coagulase negative
Staphylococci. Accordingly, there is a need in the art to provide monoclonal antibodies that can bind to Staphylococcus of both coagulase types and that can enhance phagocytosis and killing of the bacteria and thereby enhance protection in vivo
There is also a need in the art for the epitope of the site to which such antibodies can bind so that other antibodies with similar abilities can be identified and isolated
There is a related need in the art for humanized or other chimeric human/mouse monoclonal antibodies In recent well publicized studies, patients
administered murine anti-TNF (tumor necrosis factor) monoclonal antibodies
developed anti-muπne antibody responses to the administered antibody (Exley A R , et al , Lancet 335 1275-1277 (1990)) This type of immune response to the treatment regimen, commonly referred to as the HAMA response, decreases the
effectiveness of the treatment and may even render the treatment completely ineffective Humanized or chimeric human/mouse monoclonal antibodies have been shown to significantly decrease the HAMA response and to increase the therapeutic effectiveness See LoBug o et al , P N A S 86 4220-4224 (June 1989)
SUMMARY OF THE INVENTION
To address these needs in the art, the present invention encompasses opsonic and protective monoclonal and chimeric antibodies that bind to lipoteichoic
acid of Gram positive bacteria The antibodies also bind to whole bacteria and
enhance phagocytosis and killing of the bacteria in vitro and enhance protection
from lethal infection in vivo The mouse monoclonal antibody has been humanized and the resulting chimeric antibody provides a previously unknown means to
diagnose, prevent and/or treat infections caused by gram positive bacteria bearing lipoteichoic acids. This invention also encompasses a peptide mimic of the lipoteichoic acid epitope binding site defined by the monoclonal antibody. This
epitope or epitope peptide mimic identifies other antibodies that may bind to the lipoteichoic acid epitope. Moreover, the epitope or epitope peptide mimic provides a valuable substrate for the generation of vaccines or other therapeutics.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 provides a schematic representation of lipoteichoic acid (LTA) in the Gram positive bacterial cell wall.
Figure 2 depicts antibody regions, such as the heavy chain constant region
(CH), the heavy chain variable region (VH), the light chain constant region (CL), and the light chain variable region (VL).
Figure 3 demonstrates the enhancement of survival after administration of MAB 96-110 in a lethal neonatal model of coagulase positive staphylococcus sepsis.
Figure 4 demonstrates enhancement of survival in adult mice infected with coagulase negative staphylococci after administration of MAB 96-110.
Approximately 23 hours after infection, 70% of the animals treated with MAB 96-110 were alive compared with 20% of animals not given antibody.
Figure 5 (SEQ ID NOS 4 & 5, and 6 & 7, respectively) provides a list of 18
resulting sequences for the 6mer library panning.
Figure 6 (SEQ ID NOS 8-43, respectively) provides a list of the 18 resulting
sequences for the second experiment 15mer library panning. Figure 7 (SEQ ID NOS 44 & 45, and 46 & 47, and 48 & 49, respectively) provides a list of the 17 resulting sequences for the first experiment 15mer library
panning.
Figure 8 (SEQ ID NOS 50-67, respectively) provides a master list compiled from the common resulting peptide sequences from all the pannings.
Figure 9 sets forth a comparison of the optical density signals of each phage
isolate at 6.25 x 1011 vir/ml.
Figure 10 shows the strategy for cloning the variable region gene fragments.
Figure 11 (SEQ ID NOS 68-85, respectively) lists the oligonucleotides primers
used.
Figure 12 (SEQ ID NOS 86-105, respectively) provides the final consensus DNA sequence of the heavy and light chain variable regions.
Figure 13 demonstrates the re-amplification of the variable region gene
fragments.
Figure 14 sets forth the plasmid map for pJRS334.
Figure 15 provides the results of the antibody production assay, showing that the transfection of cells with the plasmid construct results in the production of a molecule containing both human IgG and kappa domains.
Figure 16 provides the results of the activity assay, demonstrating that the
transfection of cells with the plasmid construct results in the production of a molecule
that binds to the Hay antigen.
Figure 17 depicts the opsonic activity of the chimeric monoclonal antibody 96-
110 for S. epidermidis in a neutrophil mediated opsonophagocytic bactericidal
assay. Figure 18 demonstrates the enhancement of survival after administration of
MAB 96-110 in a lethal model of S. epidermidis sepsis.
Figure 19 depicts the effect of the chimeric monoclonal antibody 96-110 on the survival of adult mice after intraperitoneal challenge with S. epidermidis.
Figure 20 sets forth the effect of the chimeric monoclonal antibody 96-110 on
bacteremia in a lethal S. epidermidis model.
Figure 21 depicts bacteremia levels four hours after infection with S. epidermidis at different doses of the chimeric monoclonal antibody 96-110.
Figure 22 sets forth the effect of the chimeric monoclonal antibody 96-110 on
survival in a lethal neonatal S. epidermidis sepsis model.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides anti-lipoteichoic acid (LTA) murine antibodies (including monoclonal antibodies) and chimeric murine-human antibodies, and
fragments, derivatives, and regions thereof, which bind to and opsonize whole Gram positive cocci such as Staphylococcus to thereby enhance phagocytosis and killing of such bacteria in vitro and which enhance protection from lethal infection of such bacteria in vivo. The antibodies, fragments, regions, and derivatives thereof of the
invention preferably recognize and bind to an epitope of LTA that can block the
binding of Gram positive bacteria to epithelial cells, such as human epithelial cells.
Accordingly, the invention provides broadly reactive and opsonic antibodies for the diagnosis, prevention, and/or treatment of bacterial infections caused by Gram
positive bacteria. The antibodies of the invention are broadly reactive with Gram positive bacteria, meaning that they selectively recognize and bind to Gram positive bacteria and do not recognize or bind to Gram negative bacteria. Any conventional binding assay can be used to assess this binding, including for example, the enzyme linked
immunosorbent assay described below. The basis of the binding is the presence of
LTA exposed on the surface of the cell wall of Gram positive bacteria.
As noted above, the cell walls of Gram positive bacteria characteristically contain peptidoglycans such as murein as well as teichoic acids. Teichoic acids are polymers of either giycerol phosphate or ribitol phosphate with various sugars,
amino sugars, and amino acids as substituents. Although the lengths of the chains and the nature and location of the substituents vary from species to species and sometimes between species, in general teichoic acids make up a major part of the cell wall. The teichoic acids related to this invention are lipoteichoic acids which are
teichoic acids made up of giycerol phosphate which is primarily linked to a glycolipid in the underlying cell membrane. Although the precise structure of LTA in the Gram positive bacterial cell wall is not known, a standard schematic representation commonly accepted in the art is set forth in Figure 1. Accordingly, the antibodies of the claimed invention are broadly reactive because they recognize and bind to the
lipoteichoic acids that are characteristically surface exposed on Gram positive bacteria.
The antibodies of the invention are also opsonic, or exhibit opsonic activity,
for Gram positive bacteria. As those in the art recognize, "opsonic activity" refers to
the ability of an opsonin (generally either an antibody or the serum factor C3b) to
bind to an antigen to promote attachment of the antigen to the phagocyte and thereby enhance phagocytosis. Certain bacteria, especially encapsulated bacteria which resist phagocytosis due to the presence of the capsule, become extremely attractive to phagocytes such as neutrophils and macrophages when coated with an opsonic antibody and their rate of clearance from the bloodstream is strikingly
enhanced. Opsonic activity may be measured in any conventional manner as described below.
The ability of the anti-LTA antibodies of the invention to bind to and opsonize Gram positive bacteria and thereby enhance phagocytosis and cell killing in vitro and
to enhance protection in vivo is completely unexpected because anti-LTA antibodies have been reported to lack opsonic activity. Indeed, anti-LTA antibodies have been often used as controls.
For example, Fattom et al., J. Clin. Micro. 30(12):3270-3273 (1992) examined
the opsonic activity of antibodies induced against type specific capsular polysaccharide of S. epidermidis, using as controls antibodies induced against techoic acids and against S. hominus. While type-specific antibodies were highly opsonic, anti-techoic acid antibodies were not different from the anti-S. hominus
antibodies.
Similarly, in Kojima et al., J. Infect. Dis. 162:435-441 (1990), the authors assessed the protective effects of antibody to capsular polysaccharide/adhesion against catheter-related bacteremia due to coagulase negative Staphylococci and
specifically used a strain of S. epidermidis that expresses teichoic acid as a control.
See page 436, Materials and Methods, left column, first right column, third In a
later study, the authors reached a more explicit conclusion against the utility of anti- techoic antibodies: Immunization protocols designed to elicit antibody to techoic acid but not to PS/A afforded no protection against bacteremia or endocarditis.
Takeda, et al., Circulation 86(6):2539-2546 (1991).
Contrary to the prevailing view in the field, the broadly reactive opsonic
antibodies against the LTA of Gram positive bacteria, including S. aureus and S. epidermidis, of the invention satisfy a clear need in the art. As described in the background section, both S. aureus and S. epidermidis are common causes of post¬
operative wound infections; S. epidermidis has become a major cause of nosocomial infections in patients whose treatments include the placement of foreign objects; S. epidermidis has become a common cause of peritonitis in patients with continuous ambulatory peritoneal dialysis; and S. epidermidis is now recognized as a common
cause of neonatal sepsis.
Indeed, our laboratory has recently focused tremendous efforts to find broadly opsonic antibodies as detailed in a recent series of four related applications and one issued patent, specifically:
USSN 08/296,133, filed August 26, 1994, of Gerald W. Fischer, entitled "Directed Human Immune Globulin for the Prevention of Staphylococcal Infections;"
U.S. Patent No. 5,571 ,511 , issued November 5, 1996 to Gerald W. Fischer, entitled "Broadly Reactive Opsonic Antibodies that React with Common Staphylococcal Antigens;"
USSN 08/466,059, filed June 6, 1995, of Gerald W. Fischer, entitled "In Vitro Methods for Identifying Pathogenic Staphylococcus, For Identifying Immunoglobulin Useful for the Treatment of Pathogenic Staphylococcus Infections, and In Vitro Methods Employing such Immunoglobulins;" and USSN 08/308,495, filed September 21,1994, of to Gerald W. Fischer, entitled "Broadly Reactive Opsonic Antibodies that React with Common Staphylococcal Antigens,"
all of which are specifically incorporated herein by reference.
This series of applications and the issued patent describe the search for broadly reactive opsonic antibodies particularly against Staphylococci. In rough chronological order, the "Directed Human Immune Globulin" application describes
the selection and use of Directed Human Immune Globulin to prevent or treat infections caused by S. epidermidis which contains antibodies with the ability to bind to surface antigens of S. epidermidis in an ELISA and the exhibition of greater than 80% opsonophagocytic bactericidal activity against S. epidermidis in a particularly described in vitro assay. The issued patent claims describes for the first time a particular strain of S. epidermidis that identifies broadly reactive opsonic antibodies
against both coagulase positive and coagulase negative Staphylococci and specifically claims an antigen preparation isolated from S. epidermidis strain Hay ATCC 55133, deposited on December 19, 1990, which generates broadly reactive
opsonic antibody which specifically reacts in an assay with S. epidermidis serotypes I, II and III, and which exhibits opsonic activity greater than 70%. The In Vitro Methods" application describes the use of a Serotype II S. epidermidis, such as the Hay strain, that identifies pathogenic Staphylococcus infections. The fourth
application in the chain describes a surface protein identified on the Hay strain that
can induce broadly reactive opsonic antibodies.
Nonetheless, the search continued for antibodies, both polyclonal and monoclonal, that are broadly reactive and opsonic against all Gram positive bacteria
and has culminated in the present invention. Having discovered the Hay strain and determined its unique ability to generate broadly opsonic antibodies against Staphylococci, it was used as the basis for this search.
As set forth in Example 1 , mice were immunized with whole strain Hay S. epidermidis from which hybridomas were produced. In screening the hybridomas for antibodies, the antibodies of one clone (first designated 96-105CE11 IF6 and later designated 96-110 MAB) exhibited a strong IgG reaction (Tables 1 and 2) and, in
further tests, was found to bind very strongly to Gram positive bacteria such as to
strain Hay, to all three serotypes of S. epidermidis, to S. hemolyticus, S. hominus, and two serotypes of S. aureus (Tables 3-6) but not to the Gram negative control, Haemophiius influenza.
Similar to the antibodies described in the Fischer applications and patent set
forth above, the antibody of the present invention exhibits very strong binding, i.e., O.D.s of around twice background in an enzyme-linked immunosorbent assay (described below), against strain Hay. In a preferred embodiment, the level of high binding is equal to or greater than five times background. In other embodiments, the
level of high binding is equal to or greater than 10 times background. Of course, any meaningful increase over background (the level observed when all the reagents other than the antibody being tested) will be recognized by skilled persons in the art as high binding and therefor within the scope of the invention.
Also as described in the Fischer applications and patent, high binding has
been found to correlate with opsonic activity. As set forth in Example 2, in a neutrophil mediated bactericidal assay (described below), the 96-110 MAB exhibited
enhanced opsonization against the prototypic coagulase negative bacteria, S.
epidermidis, and against the prototypic coagulase positive bacteria, S. aureus. With this level of opsonic activity, an antibody should enhance phagocytosis and cell
killing of both coagulase negative and coagulase positive bacteria.
The term "enhanced" refers to activity that measurably exceeds background at a valuable level. The level deemed valuable may well vary depending on the specific circumstances of the infection, including the type of bacteria and the severity of the infection. For example, for enhanced opsonic or phagocytic activity, in a
preferred embodiment, an enhanced response is equal to or greater than 75% over background. In another preferred embodiment, the enhanced response is equal to or greater than 80% over background. In yet another embodiment, the enhanced response is equal to or greater than 90% over background.
To confirm that the antibody, previously shown to be broadly reactive as well as opsonic, would be protective in vivo, MAB 96-110 was assessed in a lethal infection model in both neonatal rats and adult mice. As set forth in Example 3, survival in control animals given either no therapy, saline, or control MAB, ranged
from 0 to less than 10%. However, MAB 96-110 enhanced the survival to 50% or
greater.
Where, as here, the enhancement measured is of survival, the preferred increase over background may be more modest than above. Thus, an increase in
survival of 25% may be an enhanced response. In other embodiments, enhanced
survival may be greater than 50%. Again, the person of skill in the art would recognize other meaningful increases in survival as within the invention.
In view of the impressive opsonic activity in vitro as well as the protective
activity in vivo of MAB 96-110, we sought the identity of the epitope of the antigen to
which it bound. An "antigen" is a molecule or a portion of a molecule capable of being bound by an antibody and which is also capable of inducing an animal to produce antibody capable of binding to an epitope of that antigen. An antigen may have one or more epitopes. An "epitope" analogously means that portion of the molecule that is capable of being recognized by and bound by an antibody. In general, epitopes consist of chemically active surface groupings of molecules such
as amino acids or sugar side chains that have specified three dimensional structural and specific charge characteristics.
In a series of panning experiments set forth in Examples 4-6, we identified peptide sequences to which MAB 96-110 bound strongly. These sequences provide
at least peptide mimics of the epitope to which MAB 96-110 bound. Thus, one aspect of the present invention involves a peptide having the sequence
W R M Y F S H R H A H L R S P (SEQ ID NO 1) and another aspect of the invention involves a peptide having the sequence
W H W R H R I P L Q L A A G R (SEQ ID NO 2). Of course, the epitope of the invention may be identical to one of these sequences or may be substantially homologous to these sequences such that the anti-LTA antibodies of the invention will bind to them. Alternatively, the substantially homologous sequences of the invention are those that are able to induce the anti-
LTA antibodies of the invention. Other peptide epitope mimics within the invention may vary in length and sequence from these two peptides.
The present invention also encompasses recombinant epitopes, epitope
mimics, and antigens. The DNA sequence of the gene coding for the isolated
antigen can be identified, isolated, cloned, and transferred to a prokaryotic or
eukaryotic cell for expression by procedures well-known in the art. For example, procedures are generally described in Sambrook et al., Molecular Cloning, A
Laboratory Manual, 2nd Ed., Cold Springs Harbor Press, Cold Spring Harbor, New York (1989) incorporated by reference.
To confirm the specificity of the peptides for the monoclonal antibody, it was
tested in a competitive inhibition assay and found to inhibit binding of MAB 96-110 to strain Hay. See Example 6.
To determine the protein of which such sequences are a part, we compared the peptide sequences to the sequences of proteins but, as set forth in Example 7,
failed to identify any known protein. Accordingly, we expanded our search of other antigen candidates. Because the peptide sequence was small and had successfully inhibited the binding of MAB 96-110 to strain Hay and because MAB 96-110 bound to and opsonized all three serotypes of S. epidermidis as well as to both coagulase negative and coagulase positive bacteria, we assessed the possibility that the peptide was part of a surface exposed lipoteichoic acid. To our surprise, as set forth in Example 7, we found that MAB 96-110 bound to the LTAs of several Gram positive bacteria such as S. mutans. S. aureus, S. faecalis, S. pyogenes (group A Streptococcus).
Thus, the present invention includes antibodies that are capable of binding to the LTA of Gram positive bacteria, including both coagulase negative and coagulase positive bacteria, and of enhancing the opsonization of such bacteria. These anti-
LTA antibodies include polyclonal antibodies as well as monoclonal antibodies
produced by the hybridomas of the invention, such as MAB 96-110 as well as other
monoclonal antibodies, fragments and regions thereof, as well as derivatives thereof. As set forth above, the strength of the binding may range from twice above background, to five- and ten-times above background. In addition, the antibodies, fragments, regions, and derivatives of the present invention are capable of enhancing the opsonization of such bacteria, at rates ranging from 75% and up.
The "fragments" of the antibodies of the invention include, for example, Fab,
Fab', F(ab')2, and SFv. These fragments are produced from intact antibodies using methods well known in the art such as, for example, proteolytic cleavage with enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2). The "regions" of the antibodies of the present invention include a heavy chain
constant region (Hc or CH), a heavy chain variable region (Hv or VH), a light chain constant region (Lc or CL), and a light chain variable region (Lv or VL) (Figure 2). The light chains may be either a lambda or a kappa chain.
In a preferred aspect of the invention, the regions include at least one heavy
chain variable region or a light chain variable region which binds a portion of LTA, including for example the specific antigen binding sites (that which binds to the epitope) of the two regions. In another embodiment, these two variable regions can be linked together as a single chain antibody. While a full length heavy chain may
be critical for opsonic activity and enhance anti-cytokine (anti-inflammatory) activity, the antibody fragments encompassing the variable regions may be suitable for inhibition of bacterial binding to epithelial cells and may also be anti-inflammatory.
In a particularly preferred aspect of the invention, the antibody is a chimeric
mouse/human antibody made up of regions from the anti-LTA antibodies of the
invention together with regions of human antibodies. For example, a chimeric H
chain can comprise the antigen binding region of the heavy chain variable region of the anti-LTA antibody of the invention linked to at least a portion of a human heavy chain constant region. This humanized or chimeric heavy chain may be combined with a chimeric L chain that comprises the antigen binding region of the light chain variable region of the anti-LTA antibody linked to at least a portion of the human light
chain constant region.
The chimeric antibodies of the invention may be monovalent, divalent, or polyvalent immunoglobulins. For example, a monovalent chimeric antibody is a dimer (HL) formed by a chimeric H chain associated through disulfide bridges with a
chimeric L chain, as noted above. A divalent chimeric antibody is a tetramer (H2 L2) formed by two HL dimers associated through at least one disulfide bridge. A polyvalent chimeric antibody is based on an aggregation of chains.
A particularly preferred chimeric antibody of the invention is described in
Examples 8-10 which set forth in detail the preparation of a preferred chimeric IgG antibody (and in Examples 11-13 which describe the functional activity of this preferred chimeric anibody). Of course, other chimeric antibodies composed of different sections of the anti-LTA antibodies of the invention are within the invention. In particular, the heavy chain constant region can be an IgM or IgA antibody.
In addition to the protein fragments and regions of the antibodies, the present
invention also encompasses the DNA sequence of the gene coding for the antibodies as well as the peptides encoded by the DNA. Particularly preferred DNA and peptide sequences are set forth in Figure 12. That figure provides the variable
regions of both the heavy and light chains of MAB 96-110, including the
Complementarity Determining Regions ("CDR"), the hypervariable amino acid sequences within antibody variable regions which interact with amino acids on the
complementary antigen. The invention includes these DNA and peptide sequences as well as DNA and peptide sequences that are homologous to these sequences. In a preferred embodiment, these sequences are 70 % homologous although other
preferred embodiments include sequences that are 75%, 80%, 85%, 90%, and 95%
homologous. Determining these levels of homology for both the DNA and peptide sequence is well within the routine skill of those in the art.
The DNA sequences of the invention can be identified, isolated, cloned, and
transferred to a prokaryotic or eukaryotic cell for expression by procedures
well-known in the art. Such procedures are generally described in Sambrook et al., supra, as well as Current Protocols in Molecular Biology (Ausubel et al., eds., John Wiley & Sons, 1989), incorporated by reference. In one preferred embodiment, the CDR can be graphed onto any human antibody frame using techniques standard in the art, in such a manner that the CDR maintains the same binding specificity as in
the intact antibody.
In addition, the DNA and peptide sequences of the antibodies of the invention, including both monoclonal and chimeric antibodies, may form the basis of
antibody "derivatives," which include, for example, the proteins or peptides encoded by truncated or modified genes. Such proteins or peptides may function similarly to the antibodies of the invention. Other modifications, such as the addition of other sequences that may enhance the effector function, which includes phagocytosis
and/or killing of the bacteria, are also within the present invention.
The present invention also discloses a pharmaceutical composition comprising the anti-LTA antibodies, whether polyclonal, monoclonal or chimeric, as
well as fragments, regions, and derivatives thereof, together with a pharmaceutically
acceptable carrier. The pharmaceutical compositions of the invention may alter- natively comprise the isolated antigen, epitope, or portions thereof, together with a pharmaceutically acceptable carrier.
Pharmaceutically acceptable carriers can be sterile liquids, such as water, oils, including petroleum oil, animal oil, vegetable oil, peanut oil, soybean oil, mineral oil, sesame oil, and the like. With intravenous administration, water is a preferred carrier. Saline solutions, aqueous dextrose, and giycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, 18th Edition (A. Gennaro, ed., Mack Pub., Easton, Pa., 1990), incorporated by ref¬
erence.
Finally, the present invention provides methods for treating a patient infected with, or suspected of being infected with, a Gram positive bacteria such as a staphylococcal organism. The method comprises administering a therapeutically effective amount of a pharmaceutical composition comprising the anti-LTA immunoglobulin (whether polyclonal or monoclonal or chimeric, including fragments, regions, and derivatives thereof) and a pharmaceutically acceptable carrier. A patient can be a human or other mammal, such as a dog, cat, cow, sheep, pig, or goat. The patient is preferably a human.
A therapeutically effective amount is an amount reasonably believed to
provide some measure of relief or assistance in the treatment of the infection. Such
therapy as above or as described below may be primary or supplemental to additional treatment, such as antibiotic therapy, for a staphylococcal infection, an
infection caused by a different agent, or an unrelated disease. Indeed, combination
therapy with other antibodies is expressly contemplated within the invention. A further embodiment of the present invention is a method of preventing such
infections, comprising administering a prophyiactically effective amount of a pharmaceutical composition comprising the anti-LTA antibody (whether polyclonal or monoclonal or chimeric, including fragments, regions, and derivatives thereof) and a pharmaceutically acceptable carrier.
A prophyiactically effective amount is an amount reasonably believed to
provide some measure of prevention of infection by Gram positive bacteria. Such - therapy as above or as described below may be primary or supplemental to additional treatment, such as antibiotic therapy, for a staphylococcal infection, an infection caused by a different agent, or an unrelated disease. Indeed, combination therapy with other antibodies is expressly contemplated within the invention.
In another embodiment, the peptide which mimics the LTA epitope would be useful to prevent binding of Gram positive bacteria to epithelial cells and thereby inhibit colonization. For example, a pharmaceutical composition containing such a
peptide may be administered intranasally to prevent an infection or to minimize a current infection.
Yet another preferred embodiment of the present invention is a vaccine comprising the epitope, epitope mimic, or other part of the LTA antigen and a pharmaceutically acceptable carrier. Upon introduction into a host, the vaccine
generates an antibody broadly protective and opsonic against infection by Gram positive bacteria. The vaccine may include the epitope, an epitope mimic, any
mixture of epitopes and epitope mimics, the antigen, different antigens, or any
combination of epitopes, epitope mimics and antigens. Vaccinations are particularly beneficial for individuals known to be or suspected of being at risk of infection by Gram positive bacteria. This includes patients receiving body implants, such as valves, patients with indwelling catheters, patients preparing to undergo surgery involving breakage or damage of skin or mucosal tissue, certain health care workers, and patients expected to develop
impaired immune systems from some form of therapy, such as chemotherapy or
radiation therapy.
Treatment comprises administering the pharmaceutical composition (including antibodies and vaccines) by intravenous, intraperitoneal, intracorporeal injection, intra-articular, intraventricular, intrathecal, intramuscular, subcutaneous, intranasally, intravaginally, orally, or by any other effective method of administration. The composition may also be given locally, such as by injection to the particular area infected, either intramuscularly or subcutaneously. Administration can comprise
administering the pharmaceutical composition by swabbing, immersing, soaking, or wiping directly to a patient. The treatment can also be applied to objects to be placed within a patient, such as dwelling catheters, cardiac values, cerebrospinal fluid shunts, joint prostheses, other implants into the body, or any other objects, instruments, or appliances at risk of becoming infected with a Gram positive
bacteria, or at risk of introducing such an infection into a patient.
As a particularly valuable corollary of treatment with the compositions of the invention (including all anti-LTA antibodies (whether polyclonal or monoclonal or
chimeric, including fragments, regions, and derivatives thereof), all pharmaceutical
compositions based on such antibodies, as well as on epitope, epitope mimics, or
other part of the LTA antigen and vaccines based on such epitope or antigens) is the reduction in cytokine release that results from the introduction of the LTA of a Gram positive bacteria. As is now recognized in the art, LTA induces cytokines, including for example tumor necrosis factor alpha, Interleukin 6, and interferon gamma. See Takada et al., Infection and Immunity, 63 (1):57-65 (January 1995). Accordingly, the
compositions of the invention may enhance protection at three levels: (1) by binding to LTA on the bacteria and thereby blocking the initial binding to epithelial cells and preventing subsequent invasion of the bacteria; (2) by enhancing opsonization of the bacteria and thereby enhancing clearance of the bacteria from tissues and blood;
and/or (3) by binding to LTA and partially or fully blocking cytokine release and modulating the inflammatory responses to prevent shock and tissue destruction.
Having generally described the invention, it is clear that the invention overcomes some of the potentially serious problems described in the Background regarding the development of antibiotic resistant Gram positive bacteria. As set forth above, Staphylococci and streptococci (such as S. faecalis) have become increasingly resistant and, with the recent spread of vancomycin resistant strains, antibiotic therapy may become totally ineffective.
Particular aspects of the invention are now presented in the form of the following "Materials and Methods" as well as the specific Examples. Of course, these are included only for purposes of illustration and are not intended to be limiting of the present invention. MATERIALS AND METHODS
Bacteria
S. epidermidis, strain Hay, was deposited at the ATCC on December 19, 1990 under Accession No. 55133.
Hybridoma
Hybridoma 96-110 was deposited at the ATCC on June 13, 1997 under Accession No. HB-12368.
Isotvpe Determination Assay
Isotype was determined using an isotype kit obtained from Zymed Laboratories. The kit can be ordered under number 90-6550.
Binding Assays
In the binding assay of the invention, immunoglobulin is reacted with a
preparation of a Staphylococcal organism. The binding assay is preferably an enzyme-linked immunosorbent assay (ELISA) or a radioimmunoassay (RIA), but may also be an agglutination assay, a coagglutination assay, a colorimetric assay, a fluorescent binding assay, or any other suitable binding assay that is known in the
art. The assay can be performed by competitive or noncompetitive procedures with results determined directly or indirectly.
The Staphylococcus preparation may be fixed to a suitable solid support,
such as a glass or plastic plate, well, bead, micro-bead, paddle, propeller, or stick.
The solid support is preferably a titration plate. The fixed preparation is incubated
with immunoglobulin, which is isolated or in a biological fluid such as ascites, and
the amount of binding determined. A positive reaction occurs when the amount of binding observed for the test sample is greater than the amount of binding for a negative control. A negative control is any sample known not to contain antigen- specific immunoglobulin. Positive binding may be determined from a simple
positive/negative reaction or from the calculation of a series of reactions. This series may include samples containing measured amounts of immunoglobulin that specifically bind to the fixed antigen, creating a standard curve from which the amount of antigen-specific immunoglobulin in an unknown sample can be determined. Alternatively, antibody can be fixed to a solid support and im¬
munoglobulin identified by its ability to bind a bacterial preparation bound to the fixed antibodies.
The specific of the assays used in the Examples are set forth below: Immunoassay on Methanol-Fixed Bacterial: Heat-killed bacteria were
suspended in normal saline at an OD 650=0.600. Bacteria in 5 mis of the suspension were pelleted by cent fugation (approximately 1800 X g, 15 minutes, 10-15°C). The supernatant was discarded and the pellet resuspended into 12 mis of methanol (MeOH). One hundred microliters of the suspension in MeOH was distributed into
each well of Nunc Maxisorp Stripwells. The MeOH was allowed to evaporate, fixing the bacteria to the plastic. The bacteria-coated stripwells were stored in plastic bags and used within 2 months of preparation.
For evaluation of antibodies, the bacteria-coated plates were washed once
with PBS and non-specific reactive sites on the bacteria were blocked by the
addition of 120 ul/well of a solution of 1% bovine serum albumin (BSA) in PBS. After a 30-60 minute incubation, the wells were washed four times with PBS containing
0.05% Tween-20 (PBS-T). Antibody, diluted in PBS-T, was then added to the wells.
Supematants were tested at a dilution of 1 :2. Ascites and purified antibody were tested at dilutions indicated in the Tables. After addition of the antibody, the wells were incubated at room temperature for 30-60 minutes in a draft-free environment.
The wells were again washed four times with PBS-T and each well received 95 ul of detection antibody: rabbit anti-mouse IgG, conjugated to horse radish peroxidase (HRP) and diluted 1 :4000 in PBS-T. The detection antibodies were specific for mouse gamma, mu or alpha heavy chains (Zymed catalog numbers 61-6020, 61- 6820 or 61-6720 respectively), as indicated in the Tables.
Following another 30-60 incubation at room temperature, the wells were washed four times with PBS-T and each well received 100 ul of one-component TMB substrate solution (Kirkegaard and Perry Labs catalog number 50-76-05). The wells were incubated in the dark at room temperature for 15 minutes. The reaction was stopped by the addition of 80 ul of TMB stop solution (Kirkegaard and Perry Labs catalog number 50-85-05) and the absorbance of each well at 450 nm was determined using a Molecular Devices Vmax plate reader.
Immunoassay on LTA's: Reactivity of MAB 96-110 was measured by
immunoassay on wells coated with LTA's. LTA's were obtained from Sigma Chemical Company and diluted in PBS to 1 ug/ml. One hundred microliters of the 1 ug/ml solution was distributed into replicate Nunc Maxisorp Stripwells. The LTA was incubated in the wells overnight at room temperature. The unbound material was
removed from the wells with four washes of PBS-T. The wells were not blocked with
BSA or other proteins. Antibody, diluted in PBS-T, was then added to the wells and the assay continued as described above.
Competitive Inhibition of Antibody of LTA: In order to determine the ability
of LTA to inhibit binding of MAB 96-110 to wells coated with MeOH-fixed Strain Hay, a competitive inhibition assay was performed. Wells were coated in MeOH with
Strain Hay and blocked with BSA as described above. Fifty ul of LTA from S. mutans, S. aureus or S. facecalis were added to duplicate wells. Six different concentrations of each LTA were tested (from 0.04 to 9.0 ug/ml). LTA's were diluted in PBS-T to obtain the desired concentrations. Immediately after addition of the LTA, 50 ul of purified MAB 96-110 at 1 ug/ml was added to each well. The final
dilution of the MAB 96-110 was therefore 0.5 ug/ml. Uninhibited control wells received only PBS-T and MAB without LTA.
Binding of MAB 96-110 to the LTA in the PBS-T solution resulted in a
complex of MAB/LTA which was removed from the plate during the subsequent washing step. The interaction of the MAB 96-110 with the LTA inhibited the antibody from binding to the LTA on the surface of the bacteria and thus reduced the number of MAB 96-110 molecules bound to the MeOH-fixed strain Hay used to coat the wells. Because the number of MAB 96-110 molecules bound to the MeOH-fixed
Strain Hay was reduced, the level of binding of the detection antibody (rabbit anti- mouse IgG-HPR) was therefore also decreased, leading to a reduction of color development when compared to wells in which no LTA was present.
Immunoassay with Protein A Method: In order to evaluate monoclonal
antibody 96-110 for reactivity with S. aureus 5 and S. aureus 8, it was necessary to
modify the immunoassay procedure described above. Both S. aureus strains express Protein A on their surfaces. Because Protein A binds strongly to the constant region of the heavy chains of gamma-globulins, it was possible that false
positive results would be obtained due to non-specific binding of the 96-110 antibody
to the Protein A molecule. In order to overcome this difficulty, the immunoassay wells were coated with bacteria as described above. However, prior to the addition of the 96-110 antibody to the bacteria-coated wells, the purified monoclonal antibody (MAb) was reacted with a solution of recombinant Protein A conjugated to HRP and diluted 1 :500 in PBS-T. This reaction was allowed to proceed for 30 minutes. The
wells were washed four times with PBS-T and 100 ul of the solution of Protein A- HRP-MAb was added to the wells. The presence of the Protein A-HRP from the pretreatment prevented the MAb from binding to the Protein A on the S. aureus 5 and 8. Furthermore, the binding of the Protein A-HRP to the constant region of the heavy chain did not interfere with the antibody binding site on the MAb, thereby
allowing evaluation of the MAb on S. aureus and other bacteria.
The Protein A-HRP-MAb was allowed to react in the coated wells for 30-60 minutes at room temperature. The wells were then washed with PBS-T and TMB substrate solution was added and the assay completed as described above.
Opsonization assays
An opsonization assay can be a colorimetric assay, a chemiluminescent assay, a fluorescent or radiolabel uptake assay, a cell-mediated bactericidal assay, or any other appropriate assay known in the art which measures the opsonic
potential of a substance and identifies broadly reactive immunoglobulin. In an opsonization assay, the following are incubated together: an infectious agent, a
eukaryotic cell, and the opsonizing substance to be tested, or an opsonizing substance plus a purported opsonizing enhancing substance. Preferably, the
opsonization assay is a cell-mediated bactericidal assay. In this in vitro assay, the
following are incubated together: an infectious agent, typically a bacterium, a
phagocytic cell, and an opsonizing substance, such as immunoglobulin. Although any eukaryotic cell with phagocytic or binding ability may be used in a cell-mediated bactericidal assay, a macrophage, a monocyte, a neutrophil, or any combination of these cells, is preferred. Complement proteins may be included to promote
opsonization by both the classical and alternate pathways.
The opsonic ability of immunoglobulin is determined from the amount or number of infectious agents remaining after incubation. In a cell-mediated bactericidal assay, this is accomplished by comparing the number of surviving bacteria between two similar assays, only one of which contains the purported
opsonizing immunoglobulin. Alternatively, the opsonic ability is determined by measuring the numbers of viable organisms before and after incubation. A reduced number of bacteria after incubation in the presence of immunoglobulin indicates a positive opsonizing ability. In the cell-mediated bactericidal assay, positive
opsonization is determined by culturing the incubation mixture under appropriate bacterial growth conditions. Any significant reduction in the number of viable bacteria comparing pre- and post-incubation samples, or between samples which contain immunoglobulin and those that do not, is a positive reaction.
Clearance/protective assays
Another preferred method of identifying agents for the treatment or prevention of a infection by Gram positive bacteria employs lethal models of sepsis that measure clearance and protection. Such agents can be immunoglobulin or other
antimicrobial substances.
A particularly useful animal model comprises administering an antibody and a
Gram positive organism to an immunocompromised (e.g., an immature) animal, followed by evaluating whether the antibody reduces mortality of the animal or enhances clearance of the organism from the animal. This assay may use any immature animal, including the rabbit, the guinea pig, the mouse, the rat, or any other suitable laboratory animal. The suckling rat lethal animal model is most preferred.
Such a model can readily incorporate an infected foreign body, such as an infected catheter, to more closely mimic the clinical setting. An alternative model utilizes adult susceptible animals, such as CF1 mice.
Clearance is evaluated by determining whether the pharmaceutical composition enhances clearance of the infectious agent from the animal. This is
typically determined from a sample of biological fluid, such as blood, peritoneal fluid, or cerebrospinal fluid. The infectious agent is cultured from the biological fluid in a manner suitable for growth or identification of the surviving infectious agent. From samples of fluid taken over a period of time after treatment, one skilled in the art can determine the effect of the pharmaceutical composition on the ability of the animal to clear the infectious agent. Further data may be obtained by measuring over a period of time, preferably a period of days, survival of animals to which the pharmaceutical composition is administered. Typically, both sets of data are utilized. Results are considered positive if the pharmaceutical composition enhances
clearance or decreases mortality. In situations in which there is enhanced organism clearance, but the test animals still perish, a positive result is still indicated. EXAMPLE 1
The Production of Hybridomas and Monoclonal Antibodies
To produce monoclonal antibodies that were directed against the surface proteins of S. epidermidis and were opsonic and protective for S. epidermidis, mice were immunized with whole S. epidermidis, Strain Hay.
A suspension of heat killed S. epidermidis was adjusted to an optical density
(OD) of 0.137 at a wavelength of 650 nm when measured through a 1 centimeter
light path. Bacteria from five mis of the suspension were pelleted by centrifugation (approximately 1800 X g, 10 minutes, room temperature). The supernatant was discarded and the pellet resuspended in 0.6 mis of PBS, which was then mixed with 0.9 mis of complete Freund's adjuvant (CFA). The resulting emulsion was used as the immunogen.
Adult, female BALB/c mice, obtained from Harlan Sprague Dawley (Indianapolis, IN) were immunized subcutaneously with 0.2 mis of the immunogen described above. The mice received a booster immunization approximately two and
Vi months later with antigen prepared as described above, except that incomplete Freund's adjuvant (IFA) was used as the adjuvant instead of CFA. A final, prefusion boost was given approximately two months after that. This boost consisted of 1 ml of S. epidermidis suspension (OD650=0.137). Mice 8159 and 8160 each received an
intraperitoneal injection of 0.5 mis of the suspension. Five days later, the spleen
from mouse 8159 was removed and used for hybridoma formation.
Hybridomas were prepared by the general methods of Shulman, Wilde and Kohler Nature 276:269-270 (1978) and Bartal and Hirshaut "Current Methods in
Hybridoma Formation in Methods of Hybridoma Formation. Bartal and Heishaut, eds., Humana Press, Clifton, New Jersey (1987). A total of 2.135 X 108 spleenocytes from mouse 8159 were mixed with 2.35 X 107 SP2/0 mouse myeloma
cells (ATCC Catalog number CRL1581) and pelleted by centrifugation (400 X g, 10 minutes at room temperature) and washed in serum free medium. The supernatant was removed to near-dryness and fusion of the cell mixture was accomplished in a sterile 50 ml centrifuge conical by the addition of 1 ml of polyethylene glycol (PEG;
mw 1400; Boehringer Mannheim) over a period of 60-90 seconds. The PEG was
diluted by slow addition of serum-free medium in successive volumes of 1, 2, 4, 8, 16 and 19 mis. The hybridoma cell suspension was gently resuspended into the medium and the cells pelleted by centrifugation (500 X g, 10 minutes at room temperature). The supernatant was removed and the cells resuspended in medium RPMI 1640, supplemented with 10% heat-inactivated fetal bovine serum, 0.05 mM hypoxanthine and 16 uM thymidine (HT medium). One hundred ul of the hybridoma cells were planted into 760 wells of 96-well tissue culture plates. Eight wells (column 1 of plate A) received approximately 2.5 X 104 SP2/0 cells in 100 ul. The SP2/0 cells
served as a control for killing by the selection medium added 24 hours later.
Twenty four hours after preparation of the hybridomas, 100 ul of RPMI 1640, supplemented with 10% heat-inactivated fetal bovine serums, 0.1 mM hypoxanthine, 0.8 uM aminopterin and 32 uM thymidine (HAT medium) was added to each well.
Ninety six hours after the preparation of the hybridomas, the SP2/0 cells in
plate A, column 1 appeared to be dead, indicating that the HAT selection medium
had successfully killed the unfused SP2/0 cells.
Eleven days after the preparation of the hybridomas, supernatants from all
wells were tested by ELISA for the presence of antibodies reactive with methanol- fixed S. epidermidis. Based on the results of this preliminary assay, cells from 20
wells were transferred to a 24-well culture dish. Four days later, supernatant from
these cultures were retested by ELISA for the presence of antibodies reactive with methanol-fixed S. epidermidis. Of these supematants, one (from colony 96- 105CE11) was a strongly reactive IgG (Table 1). Two colonies (96-105FD4 and 96-
105GB5) were very weakly reactive IgG's and one colony 96-105HB10 was a weakly reactive IgM. Antibodies of the IgM isotype are not as desirable as IgG's and culture 96-105HB10 was cryopreserved and not further examined.
Cultures 96-105 CE11 , FD4 and GB5 were reanalyzed several days later and only CE11 showed a strong response (Table 2). No response was obtained with the other cell cultures, and no further experimental work was done with them.
To further test the specificity of this antibody for S. epidermidis, a whole cell ELISA with several bacteria was performed (Table 3). The antibodies from this colony bound strongly to S. epidermidis (Hay) O.D. 1.090 and to a lesser degree to Group B streptococcus (GBS), but not to H. influenzae (HIB+, with type b capsule; HIB- without typable capsule) or type 14, pneumococcus (Pn 14).
A clone from 96-105CE11 IF6 was isolated and retested and was an lgG-1 that reacted strongly with S. epidermidis (Strain Hay) in the whole cell ELISA (Table
4). This clone was then designated 96-110. To determine if 96-110 had the broad
binding characteristics we sought and would be consistent with binding to the
surface protein on S. epidermidis (Strain Hay) that bound broadly opsonic antibody, we ran a whole cell ELISA against several coagulase negative staphylococci (Table
5). Using 96-110 in Ascites fluid, strong binding at several dilutions was detected for
S. epidermidis type I, II, III, S. hemolyticus and S. hominus. In addition, 96-110 MAB was purified over a protein G column (Pharmacia).
Using a modification of the whole cell ELISA, peroxidase labeled protein A was mixed with the purified 96-110 MAB and then reacted with S. aureus type 5 (SA5) and S. aureus type 8 (SA8) obtained from ATCC at Accession Nos. 12602 and 12605, respectively. Both S. aureus serotypes reacted strongly with the 96-110
MAB (Table 6). Since, in our previous studies, we found that absorption with S. epidermidis (Strain Hay) could decrease IgG opsonic activity and opsonic antibodies raised against Hay reacted with a surface protein of Hay, we felt that this was still consistent with a MAB to the surface protein we were trying to characterize. This finding was also important since types 5 & 8, S. aureus are serotypes commonly associated with human infections. Using this protein A assay, MAB to type 14 pneumococcus did not demonstrate binding to S. aureus.
Therefore, we have identified a mouse IgG! MAB raised against S. epidermidis (Strain Hay) that binds to the surface of both coagulase negative and
coagulase positive Staphylococci of Gram positive bacteria. Such an antibody would be valuable to prevent or treat infections of Gram positive cocci by preventing attachment of bacteria to epithelial cells or foreign bodies, by enhancing opsonization and protection from infection and by reducing (down modulating) the inflammatory response. TABLE 1
Immunoassay Results, 96-105 Supernatants on Methanol-Fixed S. Hay
Colony Detection Specific Foi r:
ID G A M
PBS-F 0.070 0.080 0.050
CE11 0.788 0.065 0.056
EB5 0.079 0.065 0.053
EE5 0.084 0.069 0.055
FD4 0.089 0.067 0.059
FG4 0.087 0.065 0.065
FG8 0.090 0.060 0.062
FF9 0.095 0.062 0.059
GE4 0.074 0.067 0.059
GB5 0.155 0.077 0.078
GB6 0.073 0.062 0.053
GC6 0.069 0.062 0.052
GC9 0.076 0.062 0.053
GB10 0.075 0.064 0.102
HG2 0.195 0.067 0.059
HG3 0.079 0.066 0.060
HE4 0.076 0.073 0.065
HG4 0.077 0.101 0.061
HG5 0.077 0.062 0.058
HC8 0.083 0.064 0.057
HB10 0.070 0.064 0.223
AC4 IID10* 0.065 0.066 0.069
'Monoclonal antibody reactive with Hib protein D.
TABLE 2
Immunoassay Results, 96-105 Supernatants on Methanol-Fixed S. Hay
Colony Detection Specific For:
ID G A M
Buffer 0.052 0.045 0.045 CE11 0.933 0.049 0.046 FD4 0.073 0.054 0.051 GB5 0.050 0.040 0.036
TABLE 3
Immunoassay Results, 96-105 Supernatants on Methanol-Fixed Bacteria
Colony Detection ID Antibody Hav Hib+ Hib- GBS
Pn14
CE11 gamma-specific 1.090 0.106 0.068 0.304 0.063 FE11 gamma-specific 0.167 0.084 0.068 0.112 0.053 Buffer gamma-specific 0.048 0.075 0.056 0.070 0.053
Several colonies from 96-105 not cloned.
TABLE 4
Assay of 96-105 CE11 IF6 on Various Bacteria
Antibody Antigen Dilution Isotype Hay Pn14
PBS-T 0.072 0.064 05CE1 1 -IF6 Hay 2 lgG-1 , k 1.608 0.099
4 1.184 0.087 8 0.846 0.069 16 0.466 0.074
TABLE 5
Detection of Bacteria of Anti-Hay Monoclonal* in Whole Cell ELISA
Staph Staph Staph Staph Staph Staph
Antibody Dilution Hay Epi l Epi ll Epi III Hemmolyt Hominus
Buffer 0 056 0 063 0 066 0 055 0 058 0 074
96-110 100 1 448 2 334 1 524 1 241 1 197 0 868
Ascites
400 1 325 2 542 0 746 0 425 0 830 0422
1600 1 087 2452 0 369 0 176 0 680 0 185
6400 0 930 2 430 0 195 0 089 0602 0 110
25600 0 674 1 672 0 113 0 069 0 647 0 081
*Antι-Hay Monoclonal from unpurified ascites fluid
TABLE 6
Detection of Methanol-Fixed SA5, SA8 and S. Hay By Purified Monoclonal Anti-Hay Using Protein A
Anti-Hay ATCC ATCC USU
Dilution SA5 SA8 Hay
500 1.329 3.345 3.017
1000 1.275 2.141 2.266
2000 0.873 1.016 1.487
4000 0.333 0.491 0.951
8000 0.159 0.232 0.490
16000 0.132 0.149 0.331
Normal Mouse 0.101 0.090 0.082
1000
Buffer 0.102 0.113 0.152
Purified anti-Hay stock = 1.63 mg/ml
EXAMPLE 2
The Opsonic Activity of the Monoclonal Antibody
Antibodies which bind to an antigen may not necessarily enhance opsonization or enhance protection from infection. Therefore, a neutrophil mediated
bactericidal assay was used to determine the functional activity of antibody to S. epidermidis. Neutrophils were isolated from adult venous blood by dextran sedimentation and ficoll-hypaque density centrifugation. Washed neutrophils were added to round-bottomed wells of microtiter plates (approximately 106 cells per well) with approximately 3 x 104 mid-log phase bacteria (S. epidermidis Hay, ATCC 55133). Newborn lamb serum (10 uls), screened to assure absence of antibody to S. epidermidis, was used as a source of active complement.
Forty microliters of immunoglobulin (or serum) were added at various dilutions
and the plates were incubated at 37°C with constant, vigorous shaking. Samples of 10 uls were taken from each well at zero time and after 2 hours of incubation. Each was diluted, vigorously vortexed to disperse the bacteria, and cultured on blood agar plates overnight at 37 °C to quantitate the number of viable bacteria. Results are
presented as percent reduction in numbers of bacterial colonies observed compared to control samples.
Since the 96-110 MAB bound to both coagulase negative and coagulase positive Staphylococci, opsonic studies were performed to determine if the MAB enhanced phagocytosis and killing of both groups of staphylococci. At a 1:80
dilution, the MAB enhanced opsonization and killing of coagulase negative
Staphylococcus (S. epidermidis) to 100%, compared with 49.5% with C and PMN alone (Table 7). The coagulase positive Staphylococcus also showed enhanced phagocytosis at 1 :10 and 1 :40 dilution (83.3% and 78.9% respectively) compared
with 53.7 percent with C and PMN alone. At 1:80 dilution, the opsonic activity against S. aureus was decreased to 61%.
These data show that not only does the MAB bind to the surface of both
coagulase negative and coagulase positive Staphylococci, but that it has functional activity and can enhance phagocytosis and killing of these bacteria. Such an antibody would be capable of promoting clearance of Staphylococci that have invaded a host and would be useful therapeutic agent.
TABLE 7
Opsonic Assay
Antibody: Purified M X Hay, 96-110
Group Ab % Killed % Killed
Description Dilution S. epidermidis S. aureus
C only 0.0 0.0
PMN only 0.0 0.0
PMN + C No Ab 49.5 53.7
PMN + Ab + C 10 _ 83.3
40 - 78.9
80 100.0 61.0
EXAMPLE 3
In vivo Protective Efficacy
Opsonic antibody correlates with enhanced protection from staphylococcal infections, as set forth in the recent series of Fischer applications and issued patent described and incorporated by reference above. To further demonstrate that the MAB can enhance survival to infections with both coagulase positive and coagulase negative Staphylococci, studies were conducted using lethal infection models.
Two day old Wistar rats were injected with -106 S. aureus (type 5, ATCC 12605) subcutaneously just cephalad to the tail. Approximately 30 minutes before and 24 and 48 hours after infection, 0.2 ml MAB 96-110 (~320 ug) was given IP. Control animals were given an equal volume of saline or a control MAB not directed
against Staphylococci. All animals were observed daily for five days to determine
survival.
MAB 96-110 enhanced survival in this lethal neonatal model of coagulase positive staphylococcus sepsis (Figure 3): 8/15 survived after treatment with MAB 96-110, and 0/10 survived with Control MAB or 2/25 with saline treatment.
In a similar manner MAB 96-110 enhanced survival in adult mice infected with
coagulase negative staphylococci. Adult CF1 mice were given 0.5 ml S. epidermidis (Hay) IP (3.5 x 109 bacteria). About 24 hrs and 2 hrs before and 24 hrs post- infection, 320 ug of MAB 96-110 were given to one group of mice and compared with a second group infected in the same manner, but not treated with antibody. All
animals were followed 5 days to determine survival. Approximately 23 hours after infection, 70% of the animals treated with MAB 96-110 were alive compared with
20% of animals not given antibody (Figure 4). When the study was terminated 50%
of the MAB animals remained alive compared to only 10% of controls.
Thus, MAB 96-110 could enhance survival in lethal coagulase positive and
coagulase negative staphylococcal infections. This enhancement occurred in an adult model and an immunocompromised model (immature neonatal immune system).
EXAMPLE 4
Peptide selection Panning random 6mer and 15mer fd-tet phage libraries
Amplified random 6mer and 15mer fd-tet phage libraries were panned against
the 96-110 antibody to yield populations of 6 and 15 amino acid length peptides
which cross react with the 96-110 antibody. The original libraries were acquired from George P. Smith, Division of Biological Sciences, University of Missouri, Columbia, MO. In order to be used for panning, the 96-110 antibody was crosslinked to Biotin using the Sulfo-NHS-biotin ester crossiinking kit following the
manufacturers protocol (Pierce Chemical Co.).
For the first round of panning, 35mm polystyrene petri dishes (Costar) were coated with streptavidin by incubating the plates overnight at 4°C rocking with 100mM NaHCO3 and 10ug streptavidin. Streptavidin was then discarded and plates were filled with blocking solution (0.1 M NaHCO3, 5mg/ml dialyzed BSA, 0.1 ug/ml streptavidin) and incubated for 1hr at 4°C. The following protocol was then followed:
Wash dishes six times with TBS/Tween (50mM Tris-HCI pH 7.5, 150mM NaCl, 0.5% v/v Tween 20). Incubate dishes overnight at 4°C rocking with 400ul TBS/Tween
containing 1 mg/ml dialyzed BSA and 10ug biotinylated 96-110 antibody. Add 4ul
10mM biotin and allow to incubate 1 hr at 4°C rocking. Wash dishes six times as
previously stated. Add 400ul TBS/Tween into each dish, add 4ul 10mM biotin and
add approximately 5ul of either the 6mer or 15mer amplified fd-tet phage library (at 1x1014 vir/ml). Rock dishes 4hrs at 4°C. Pour out phage and wash ten times with TBS/Tween. Incubate plates at room temp with 400ul elution buffer (0.1 N HCl, pH adjusted to 2.2 with glycine, 1 mg/ml BSA) for 10 min with rocking. Remove eluate to a Centricon 30 (Amicon) concentrator and buffer exchange with TBS (50mM Tris-
HCI pH 7.5, 150mM NaCl) and concentrate to a volume of about 100ul. Amplify eluate by mixing 100ul eluate with 100ul K91 terrific broth ceils and allowing phage to infect cells for 10-30min. Pipette infection mixture into 20ml pre-warmed NZY
medium (10g Λ/Z amine A, 5g yeast extract, 5g NaCl, 1 liter water, adjust to pH 7.5, autoclave) containing 0.2 ug/ml tetracycline. Shake vigorously at 37°C for 30-60 min. Add 20ul of 20 mg/ml tetracycline stock to the flask. Remove a small sample for titering on plates and allow flask to shake vigorously overnight at 37°C. Calculate yield from biopanning using the number of colonies counted on the titering
plates from amplification infection and the number of input phage at the beginning of panning. This number should amount to at least approximately 10"5%. Centrifuge 20ml culture for 10min at 5,000 rpm, then for 10min at 10,000rpm; pour the doubly cleared supernatant into a fresh tube containing 3ml PEG/NaCI (16.7% PEG 8000,
3.3M NaCl). Mix well and allow to incubate overnight at 4°C. Centrifuge tube 15 min at 10,000rpm, discard supernatant and redissolve phage pellet in 1ml TBS. Collect resuspended phage into a 1.5ml eppendorf tube, clarify the suspension by centrifugation, and add 150ul PEG/NaCI. Allow to incubate on ice for 1 hr.
Microfuge the tube 10min, discard supernatant, and redissolve phage in 200ul TBS.
The second and third round of panning are carried out the same way. The
eluted, amplified phage (100ul) from the previous panning is preincubated with biotinylated 96-110 antibody (100nM for the second round; 0.1 nM for the third
round) overnight at 4°C. 400 ul TBS Tween is added to the mixture and it is pipetted onto streptavidin coated plates (prepared as previously stated) and then incubated with rocking gentle at room temperature for 10 min. The plates are then
washed, eluted, and amplified as previously stated. The input and resultant phage are titered to determine yield from biopanning.
EXAMPLE 5
Sequencing resulting phage populations to identify consensus sequences
After the third round of panning, the titered infection plates are used to pick
20 single isolated colonies for each library. The colonies are grown overnight in 5ml
NZY medium containing 40ug/ml tetracycline. Replicative form DNA is extracted
from each culture using Qiaplasmid quick prep kit (Qiagen Inc.) following the manufacturer's protocol. Media supernatants are saved for phage stock to be used in Example 4. 2.5ul of each RF DNA sample is added to a reaction containing
3.5pmole CLC502 primer (5'-TGAATTTTCTGTATGAGGTTT-3') (SEQ ID NO 3), 8 Ul
Prizm sequencing mix (ABI Inc.), QS to 20ul with water and amplified following manufacturers protocol. Successful sequences are translated and aligned. 18 resulting sequences for the 6mer library panning are listed in Figure 5. 18 resulting sequences for the second experiment 15mer library panning are listed in Figure 6.
17 resulting sequences for the first experiment 15mer library panning are listed in
Figure 7. A master list was compiled of the common resulting peptide sequences
from all the pannings (Figure 8) with the frequency of occurrence listed to the right of each sequence. Consensus portions of the sequences are marked on the master
list (Figure 8). EXAMPLE 6
Phage EIA comparing 3rd round phage isolates crossreactivity to 96-110 antibody
The saved media phage stocks for each of the common resulting peptide sequences were amplified as previously stated. Amplified phage preparations were quantitated by Abs269 and diluted to 1X1013 vir/ml and serially diluted 1.2 seven
times. A 96-well polystyrene plate was coated with 2ug/ml streptavidin in 0.1 M
NaHCO3 overnight at 4°C. Plates were emptied and blocked for 1hr at room temperature with phage blocking solution, 100ul/well. The following protocol was then followed. Wash wells three times with TBS/Tween. Incubate overnight at 4°C with 0.2ug/ml biotinylated 96110 in phage blocking solution, 100ul/well. Wash wells three times with TBS/Tween. Incubate overnight at 4°C with serially diluted phage,
100ul/well. Wash wells three times with TBS/Tween. Incubate 1 hr at room temperature with 1:5000 goat polyclonal anti-phage-HRP. Wash wells three times with TBS/Tween. Develop with 100ul ABTS substrate (Kirkegaard Perry) for
10-15min and read absorbance (402nm) on spectrophotometer according to
manufacturer's protocol. Optical density signals of each phage isolate at 6.25x1011 vir/ml are compared in Figure 9. The two isolates yielding the greatest signals are:
15mer 2.12 W R M Y F S H R H A H L R S P (SEQ ID NO:1)
15mer 2.1 W H W R H R I P L Q L A A G R (SEQ ID NO:2)
EXAMPLE 7
Antibodies Against Lipoteichoic Acid (LTA)
As set forth above, we identified two peptides that reacted with 96-110 MAB.
However, after identifying the peptides, the sequences did not correspond to any known proteins. Thus we began to consider other possible antigen candidates. We
were surprised to find that MAB 96-110 bound strongly to LTA from several gram positive bacteria such as S. mutans, S. aureus and S. faecalis (Table 8). In addition, in an ELISA, when the wells coated with S. epidermidis (Strain Hay) were reacted with MAB 96-110 inhibited by varying concentrations of LTA (from S. mutans, S.
aureus and S. faecalis), reduction in MAB binding occurred (Table 9). The inhibition of MAB 96-110 binding was greatest at the highest concentration of LTA inhibitor (9 ug/ml for each LTA) and varied according to which bacterial LTA was used (52% inhibition with S. mutans, 40.6% with S. aureus and 38.2% with S. faecalis).
The MAB 96-110 was also analyzed for binding to LTA from S. pyogenes
(group A streptococcus) and various group A streptococcal M types. The MAB showed strong binding to the LTA and also bound to the different M types with strongest binding to M1 and M3 (Table 10).
We were surprised to find an antibody that bound to LTA and enhanced
opsonization for both coagulase positive and coagulase negative staphylococci in vitro and enhanced survival in lethal models of staphylococcal (coagulase negative and coagulase positive) sepsis, in vivo. This is particularly surprising because the
bacteria in each model were injected systemically (SQ or IP) and by-passed the epithelial barriers (skin or mucous membranes) where LTA is thought to possibly act
as an adherence factor for the bacteria to epithelial ceils.
In addition, this strong anti-LTA reactivity will provide a method to block the
binding of LTA bearing bacteria to epithelial cells and prevent colonization of
important pathogens such as staphylococci, group A streptococci, S. faecalis
(enterococci) and S. mutans. Since LTA induces proinflammatory cytokines such as TNF, IL-6 and Interferon gamma, MABs with strong anti-LTA binding will also have an anti-inflammatory action and modulate cytokine production secondary to LTA bearing bacteria. Anti-LTA antibodies or vaccines could be designed and produced to modulate cytokine production and inflammation in tissues and prevent the
adverse effects of these proinflammatory cytokines.
TABLE 8 Reactivity of Anti-Hay MAB 96-110 on wells Coated with Several LTA's
Antibody ID Concentration LTA from LTA from S. LTA from S or Dilution S. mutans aureus faecalis
Buffer _ 0.145 0.172 0.140
Anti-Hay 0.9 ug/ml 3.899 3.253 3.153
MAB 0.3 ug/ml 3.523 2.824 2.769
96-110 0.1 ug/ml 2.023 2.421 2.133
0.033 ug/ml 2.143 1.590 1.539
0.011 ug/ml 1.396 0.998 0.832
TABLE 9
Inhibition of Anti-Hay MAB 96-110 with LTA From Different Gram Positive Bacteria
LTA Inhibitor LTA LTA LTA
(ug/ml) S. mutans S. aureus S. faecalis
9 0.298 0.360 0.140
3 0.449 0.434 0.496
1 0.549 0.538 0.545
0.37 0.558 0.526 0.549
0.12 0.509 0.735 0.582
0.04 0.574 0.614 0.671
0 0.621 0.607 0.648
NOTES:
1. Wells were coated with methanoi-fixed Hay.
2. Wells were blocked with 1 % BSA in PBS.
3. Monoclonal anti-Hay was used at a final concentration of 0.5 ug/ml and reacted with inhibitors at the concentrations indicated in the Table.
4. Detection was with a gamma-specific Rabbit anti-Mouse.
5. Substrate was TMB.
TABLE 10
Reactivity of MAB 96-110 on Whole Methanol Fixed Group A Strep
Antibody ID Dilution or Cone. GAS* GAS Type GAS Type GAS Type Response on Type 1 3 18 24 pyogenes LTA
#12344 #21546 #12357 #10782
Buffer 0.511 0.161 0.234 0.148 0.075
Anti-Hay MAb 0.3 ug/ml 1.377 1.113 0.844 0.566 Anti-Hay Mab 0.1 ug/ml 1.016 0.553 0.555 0.402 2.228
*AII Group A Streptococcus (GAS) from ATCC (accession numbers noted above); plates were coated with MeoH-fixed bacteria and read at 15 minutes.
EXAMPLE 8
Humanization of the Anti-Staph antibody 96-110 Cloning of the 96-110 variable region cDNAs
The hybridoma cell producing the 96-110 antibody was obtained as described above. A vial of cells was thawed, washed with serum free medium and then resuspended in IMDM (Mediatech) complete media supplemented with 10%FBS
(Irvine). Total RNA was isolated from 1 108 96-110 cells using the Midi RNA Isolation kit (Qiagen) following the manufacturer's procedure. The RNA was dissolved in 10mM Tris, 0.1 mM EDTA (pH8.4) containing 0.03U/μg Prime RNase Inhibitor (5'-3') to a final concentration of 0.25 μg/μl.
Figure 10 shows the strategy for cloning the variable region gene fragments and Figure 11 lists the oligonucleotides primers used. The 96-110 total RNA (2 μg) was converted to cDNA by using Superscript ll-MMLV Reverse transcriptase (Life Technologies) and mouse kappa (OKA57) and mouse heavy chain (JS160-162)- specific priming according of manufacturer's procedures. The first strand cDNA synthesis products were then purified using a Centricon-30 concentrator device (Amicon). Of the 40 μl of cDNA recovered, 5 μl was used as template DNA for PCR. Typical PCR amplification reactions (100 μl) contained template DNA, 50 pmoles of
the appropriate primers (PMC12-15,55 and OKA57 for tight chains, JSSI-4,8 and JS
160-162 for heavy chains), 2.5 units of ExTaq polymerase (PanVera), 1x ExTaq
reaction buffer, 200 μM dNTP, 1mM MgCI2. The template was denatured by an initial incubation at 96°C for 5 min. The products were amplified by 15 thermal
cycles of 55°C for 30 sec, 70°C for 30 sec, then 96°C for 1 min. followed by 25 step
cycles of 70°C for 1 min., then 96°C for 1 min. The PCR products from the successful reactions were purified using the Wizard PCR Purification system
(Promega) as per manufacturer's procedure.
The heavy chain PCR products (approximately 400 bp) were then cloned into a bacterial vector for DNA sequence determination. Ligations of the PCR fragments were carried out into the PCR2.1 (invitrogen) T/A style cloning vector following the
manufacturer's procedures using a 3:1 insert to vector molar ratio. One half (5 ul) of the ligation reactions were used to transform Ultracompetent XL2Blue cells (Stratagene) as per the manufacturer's procedure. Plasmid clones containing DNA inserts were identified using diagnostic restriction enzyme digestions with Ncol (New
England Biolabs). The DNA sequence of plasmids (pJRS308) containing inserts of
the appropπate size (400bp) was then determined. The final consensus DNA sequence of the heavy chain variable regions is shown in Figure 12.
The light chain PCR products were treated differently. The hybridoma cell line that expresses the 96-110 antibody was made by fusing mouse spleenocytes with
the SP20 myeloma cell line. The SP20 cell line transcribes a pseudogene for the kappa light chain. In addition, the hybridoma cell line that expresses the 96-110 antibody transcribes a second pseudogene product for a kappa light chain that apparently arose from the spleenocyte partner of the hybridoma fusion event. This
second pseudogene transcript can be expressed from an expression vector
transfected into mammalian cells, but this recombinant antibody product does not bind to heat-killed Staph HAY cells in an ELISA (see Example 9). Both of these
pseudogene transcripts, when converted to cDNA by RT-PCR, contain an Af/lll
restriction site. For this reason, the PCR products synthesized for the light chain
variable region was digested with Af/lll and those products that did not cut were then cloned into the pGEM T-Easy (Promega) T/A style cloning vector using the
manufacturer's procedures. Light chain candidate (pJRS319) clones were digested with EcoRI (New England Biolabs) using the manufacturer's procedures to identify clones containing inserts of the appropriate size (350bp). The final consensus DNA sequence of the light chain variable regions is shown in Figure 12. The amino acids encoded by these sequences match the N-terminal amino acid analyses of the heavy and light chain peptide fragments produced by the hybridoma cell line.
The heavy and light chain variable regions were then subcloned into a mammalian expression plasmid vector pSUN 15 for production of recombinant chimeric antibody molecules. The creation of the expression vector was an extensive process of DNA fragment ligations and site directed mutagenesis steps. The result was a vector that expresses both antibody chains with CMV promoter driven transcription. Neomycin resistance serves as a dominant selectable marker for transfection of mammalian cells. In addition, it has been designed to allow convenient cloning of any light chain variable region as EcoRVISs BI fragment, any heavy chain variable region as a Nrul/EcoRI fragment, and any heavy chain constant domain as an EcoRIINotl fragment. These restriction sites were chosen because
they occur rarely (if ever) in human and mouse variable regions. There is a mouse J
region/kappa intron fragment fused to a human kappa exon so that after post transcriptional splicing a mouse human chimeric kappa light chain is produced.
The backbone of the vector was the plasmid pCDNA3 (Invitrogen). This
plasmid was cut with Hindlll/Xhol and a "light chain polylinker" DNA fragment was
inserted to create the stated "light chain vector." This linker contained the restriction
sites H/n lll, Kpnl, Cla/, Pm/l, EcoRV Xmal, BamHI and Xhol to facilitate subsequent cloning steps to create the piasmid pCDNA3.LCPL. A Smal/ Bell DNA fragment containing a light chain leader, anti-CKMB kappa light chain genomic fragment, and 3' UTR was cloned into the EcoRV/BamHI sites of pCDNA3.LCPL. The mouse kappa intron, exon and 3' UTR in this fragment was derived from LCPXK2 received
from Dr. Richard Near (Near, Rl et al, 1990, Mol Immunol. 27:901-909). Mutagenesis was then performed to eliminate an Nrul (209), Mlul (229). and BstBI (2962) and to introduce an Nhel (1229) and a BamHI (1214) site to create pCDNA3mut.LCPL.LCVK.
A second "heavy chain vector" was constructed from the pCDNA3mut.
LCPL.LCVK plasmid by replacing the light chain expression region (Hindlll/Xhol) with a "heavy chain polylinker" consisting of restriction sites Hpal, BspEI, EcoRV, Kpnl, and Xhol. A Smal/ Kpnl DNA fragment contains a heavy chain leader, antiCKMB lgG2b chain genomic fragment. A Kpnl/Sall oligo nucleotide fragment containing a 3' UTR and a NotI upstream of the Sail site was subsequently cloned into the KpnUXhol digested plasmid, (knocking out the Xhol site) to create the plasmid pCDNA3mut.HCPLHCV2b.
From this point two vectors were created that did not have any of the anti-
CKMB variable or constant domain DNA sequences. This was done by cutting the plasmid pCDNA3mut.LCPL.LCVK with EcoRV/Xhol and inserting a linker oligonucleotide fragment containing EcoRV, BstBI, and Xhol sites to create pSUN9.
In a similar way, the anti-CKMB fragment in pCDNA3mut.HCPLHCV2b (Nrul/Notl)
was replaced by a linker oligonucleotide fragment containing Nrul, EcoRI and NotI
sites to create pSUNIO. A human kappa light chain constant domain was then cloned into pSUN9 as a BstB/Xhol fragment, and a human lgG1 constant domain was cloned into pSUNIO as a EcoRI/NotI fragment. A Bglll/Nhel fragment from the human heavy chain vector was then cloned into the human light chain vector cut with BamHI/Nhel to create pSUNIδ.
This vector results in the production of recombinant antibody molecules under
the control of the CMV transcriptional promoters. The heavy chain molecules are direct cDNA constructs that fuse the variable region sequence directly into the human lgG1 constant domain. The light chain molecules, on the other hand, have a mouse kappa intron region 3' of the variable region coding fragment. After splicing the variable region becomes fused to a human kappa constant region exon. The selectable marker for the vector in mammalian coils is Neomycin (G418).
The variable region gene fragments were re-amplified by PCR using primers that adapted the fragments for cloning into the expression vector (see Figures 12 and 14). The heavy chain front primer (96110HF2) includes a 5' tail that encodes the C-terminus of the heavy chain leader and an Nrul restriction site for cloning, while the heavy chain reverse primer (96110HB) adds a 3' EcoRI restriction site for cloning. The light chain front primer (96110bLF) converts the first amino acid of the
96-110 light chain variable region sequence from glutamine (Q) to aspartic acid (D)
via the introduction of an EcoRV restriction site at the N-terminus of the light chain variable region for cloning, while the light chain reverse primer (96-110bLB) adds a 3' DNA sequence for the joining region-kappa exon splice junction followed by a
6sfB1 restriction site for cloning.
Because the last amino acid of the light chain variable region is an arginine (R) which is a very rare amino acid at this position, the reverse primer introduces a
point mutation in the codon for amino acid 106 that converts it to the much more common lysine (L). This was done because the splice junction in the expression vector for the kappa chain was derived from a J region that encoded a lysine at this position. Neither mutation in the recombinant form of the antibody would be
anticipated to alter the antibodies binding characteristics.
PCRs were performed as described above except 10ng of plasmid template was used in each case. Following a 5 min. incubation at 96 °C, the PCR perimeters were 35 thermal cycles of 58°C for 30 sec, 70°C for 30 se , and 96°C for 1 min.
The 96-110 heavy chain PCR product (approximately 400 bp) was digested with Nrul and EcoRI (New England Biolabs), purified using a Qiaquick PCR Purification column (Qiagen), as described by the manufacturer, and ligated into Nrul I EcoRI digested and gel-purified pSUN15, resulting in plasmid pJRS311 (see Figure
13).
At this point a Bst l/Noti (New England Biolabs) DNA fragment containing a mouse kappa J-kappa intron fragment fused to a human kappa exon fragment was digested and gel-purified from the vector tKMC180C1. This fragment was ligated into the backbone of pJRS311 digested with BstBllNoti and gel-purified resulting in the plasmid pJRS315 (see Figure 13).
This was the plasmid into which was cloned the 96-110 light chain variable region. The 96-110 light chain PCR product (approximately 350 bp) was digested
with EcoRV and BstBI (New England Biolabs), purified using a Qiaquick PCR
Purification column (Qiagen), as described by the manufacturer, and ligated into
EcoRV/βs/BI digested and gel-purified pJRS315, resulting in plasmid pJRS326 (see Figure 13). It was determined that during this cloning process, a deletion of approximately 200bp occurred at the intron exon junction of the kappa light chain. To repair this, an identical DNA fragment (also a BstBUNoti restriction fragment) was gel-purified
from digested pEN22 and ligated into BstBl/Noti digested and gel-purified pJRS326, resulting in the final expression plasmid construct pJRS334 (see Figures 13 and 14). The sequence of the variable regions and leader and other junctions was verified
prior to mammalian cell transfection.
EXAMPLE 9
Transient production of recombinant chimeric mouse/human 96-110 antibody
Two individual clones of the plasmid pJRS334 (pJRS334-1 , -2) were
transfected into COS and CHO cells using Superfectant (Qiagen) in 6 well tissue culture cells as described by the manufacturer. After three days the supernatant was assayed for the production of "humanized" antibody and for the capability for the expressed antibody to bind to the heat-killed Staph antigen.
Antibody production assays were preformed in 8-well strips from 96-well microtiter plates (Maxisorp F8; Nunc, Inc.) coated at a 1 :500 dilution with Goat anti- Human IgG antibody (Pierce) using a bicarbonate coating buffer, pH 8.5. The plates are covered with pressure sensitive film (Falcon, Becton Dickinson) and incubated
overnight at 4°C. Plates are then washed once with Wash solution
(lmadazole/NaCI/0.4% Tween-20). 100 microliters of culture supernatant was then applied to duplicate wells and allowed to incubate for 30 minutes on plate rotator at room temperatures. The plates were washed five times with Wash solution. A Goat
anti Human kappa-HRP (Southern Biotechnologies) conjugate was diluted 1 :800 in the sample/conjugate diluent. 100 microliters was added to the samples, then incubated on a plate rotator for 30 minutes at room temperature. The samples were
washed as above and then incubated with 100 μUwell of ABTS developing substrate (Kirkgaard & Perry Laboratories) for 10-15 minutes on a plate rotator at room temperature. The reaction was stopped with 100 μLJwell of Quench buffer (Kirkgaard & Perry Laboratories) and the absorbance value at 405 nm was
determined using an automated microtiter plate ELISA reader (Ceres UV900HI, Bioteck, Winooski, Vermont). As a positive control, a humanized mouse/human chimeric antibody BC24 was used. This assay (see Figure 15) demonstrates that the transfection of cells with this plasmid construct to results in the cells producing a molecule containing both human IgG and kappa domains. The supernatants were then assayed for the ability of the expressed antibodies to bind to heat-killed Staph. The activity assays were preformed in 8-well strips from 96-well microtiter plates (Maxisorp F8; Nunc, Inc.) coated at 0.09 OD/well with heat-killed Staph Hay cell material suspended in MeOH. The plates are left uncovered and incubated
overnight at 4°C. Plates are then washed once PBS. 100 microliters of culture supernatant was then applied to duplicate wells and allowed to incubate for 60 minutes on plate rotator at room temperature. The plates were washed five times with Wash solution. The goat anti Human kappa-HRP was diluted 1 :800 in the
sample/conjugate diluent. 100 microliters was added to the samples, then incubated
on a plate rotator for 30 minutes at room temperatures. The samples were washed as above and then incubated with 100 μL/well of ABTS developing substrate
(Kirkgaard & Perry Laboratories) for 10-15 minutes on a plate rotator at room
temperature. The reaction was stopped with 100 μUwell of Quench buffer (Kirkgaard & Perry Laboratories) and the absorbance value at 405 nm was determined using an automated microtiter plate ELISA reader (Ceres UV900HI,
Bioteck, Winooski, Vermont). As a positive control, the original mouse monoclonal antibody 96-110 was used, and assayed with a Goat anti-Mouse Fc-HRP conjugate @ 1 :2000 dilution. This assay (see Figure 16) demonstrates that the transfection of
cells with this plasmid construct to results in the cells producing a molecule that
binds to the Staph Hay cellular antigen.
EXAMPLE 10
Stable production of recombinant chimeric mouse/human 96-110 antibody
The plasmid pJRS334-1 was transfected into NS/0 cells (obtainable from Baxter International) and CHO cells using electroporation. The plasmid was linearized with Pwvl restriction digestion. 25 micrograms of digested plasmid DNA
was mixed with 1x107 cells in a total volume of 800 microliters in a 4 centimeter cuvette and subjected to a pulse of 250mA, 9600microF. The cells were plated out after 24 hours in 10ml non-selective media. The cells were then diluted out into 96- well microtiter plates. As colonies appeared, the supernatants were assayed for the production of "humanized" antibody and for the capability for the expressed antibody
to bind to the heat-killed Staph antigen. Antibody production and activity assays for
the stable transfectants were performed as described above. These assays demonstrate that the transfection of cells with this plasmid construct can result in the
production of a stable cell line that produces a humanized chimeric version of the
96-110 mouse hybridoma antibody. EXAMPLE 11
Opsonic Activity
Having produced a chimeric anti-LTA MAB for staphylococci, we tested its functional activity using S. epidermidis as a representative staphylococcal organism. Using the neutrophil mediated opsonophagocytic assay described generally in the
Material and Methods section, we assessed the MAB's opsonic activity by evaluating the percent of bacteria killed after two hours of incubation.
Neutrophils, specifically polymorphonuclear neutrophils, were isolated from adult venous blood by dextran sedimentation and ficoll-hypaque density centrifugation. Washed neutrophils were added to round-bottomed wells of microtiter plates (approximately 106 cells per well) with approximately 3 x 104 mid-log phase bacteria (S. epidermidis Hay, ATCC 55133). Human sera (10 uls), screened to assure absence of antibody to S. epidermidis, was used as a source of active complement (C-Barb-Ex (1:4)).
Forty microliters of immunoglobulin were added at various concentrations (20 ug/ml, 40 ug/ml, 80 ug/ml, and 160 ug/ml) and the plates were incubated at 37°C with constant, vigorous shaking. Samples of 10 uls were taken from each well at
zero time and after 2 hours of incubation. Each was diluted, vigorously vortexed to
disperse the bacteria, and cultured on blood agar plates overnight at 37°C to
quantitate the number of viable bacteria. Results are presented in Figure 17 as percent reduction in numbers of bacterial colonies observed compared to control
samples.
Compared to PMN alone or PMN plus complement, the addition of the MAB
markedly enhanced opsonic activity for staphylococcus at 20-160 ug/ml). These data demonstrate that the MAB has functional activity and can enhance the phagocytosis and killing of staphylococcal organisms, as represented by S. epidermidis.
EXAMPLE 12
In vivo Protective Efficacy
Using the lethal staphylococcal sepsis in adult mice assay (described in Example 3), we compared protection between the original mouse MAB and the chimeric HuMAB. Adult CF1 mice were given 0.5 ml S. epidermidis (Hay) IP (3.5 x
109 bacteria). About 24 hrs and 1 hr before infection, 14 mg/kg of each MAB was given to a group of mice, with a third group of mice given only PBS. All animals were followed for 40 hours after challenge to determine survival.
As set forth in Figure 18, approximately 18 hours after infection, all the control animals died while both treatment groups exhibited 100% survival. At 30 hours after infection, both MAB treatment groups exhibited 70% survival. At the end of the study, the group that received the mouse MAB exhibited greater survival than the group receiving the chimeric MAB, but both MAB enhanced survival over the PBS controls.
We conducted further studies with the chimeric MAB at a dose of 18
mg/kg/dose 2 doses given IP 24 and 1 hour prior to infection (3x109 IP S. epidermidis, Hay). As set forth in Figure 19, the chimeric MAB enhanced survival.
We also assessed the effect of the chimeric MAB on bacteremia in the lethal
S. epidermidis sepsis model. CF-1 mice were twice infected IP with strain Hay and
the chimeric MAB. Bacteremia is expressed as the number of bacteria isolated on blood agar after a 1 : 1000 dilutions. As set forth in Figure 20, the chimeric MAB reduced bacterial levels by over 2 logs. Additional studies demonstrated that bacteremia was reduced to a greater degree using 40 mg/kg/dose compared to 20 mg/kg/dose even if survival was comparable. See Figure 21.
These data indicate that increasing the amount of antibody resulted in increased bacterial clearance in vivo. Such a response is similar to the observed enhanced opsonic activity in vitro as seen when antibody was increased from 20 ug/mg to 160 ug/ml in the neutrophil mediated opsonophagocytic assay (Figure 17).
EXAMPLE 13
In vivo Protective Efficacy
The effect of the chimeric MAB 96-110 was then analyzed in a neonatal staphylococcal model using suckling rats with a foreign body infection. Two day old
Wistar rats were treated with lipid emulsion (as is standard in newborn care for nutritional purposes) 0.2 ml, 20% IP on day -1 and again on day +1 and +2 to induce further compromise of the immuno system. In two studies, we injected approximately 5 x 107 of four different strains of S. epidermidis, identified below in
Table 11 SQ through a plastic catheter and the catheter was left in place under the skin. Saline, 0.2 ml, or MAB 96-110, 0.2 ml (dose of 50-60 mg/kg), was given IP 30 min before and 24 hours after infection. The animals were followed for 5 days.
As set forth in Table 11 , in study I, survival for animals receiving MAB ranged
from 67% to 83%, with an average of 76%, in contrast to saline treatment, which
ranged from 33% to 50%, with an average of 39%. Study II showed even more
impressive results. Survival for animals treated with MAB ranged from 83% to 100%, with 90% average, compared to the saline controls at 33% to 50%, with an average of 40%. The complied data for study II are shown in Figure 22.
TABLE 11
The Effect of Hu96-110 on Survival in a Lethal
Neonatal S. eDidermidis Sepsis Model
Litter S. epidermidis Monoclonal Antibody Saline Control Number strain Treated Survived (%) Treated Survived (%)
Study 1
31 Haywood (type II) 6 4 (67%) 6 2 (33%) (clinical)
32 35984 (type I) 5 4 (80%) 4 2 (50%) (Prototype)
33 Summer (48357) 6 4 (67%) 6 2 (33%) (clinical)
34 SE-360 (type III) 6 5 (83%) 6 2 (33%) (Prototype)
35 Haywood (type II) 6 5 (83%) 6 3 (50%) (clinical)
TOTAL 29 22 (76%) 28 11 (39%)
Study II
36 Haywood (type II) 6 6 (100%) 6 3 (50%) (clinical)
37 35984 (type I) 6 5 (83%) 6 2 (33%) (Prototype)
38 Summer (48357) 6 6 (100%) 6 2 (33%) (clinical)
39 SE-360 (type III) 6 5 (83%) 6 2 (33%) (Prototype)
40 Haywood (type II) 6 5 (83%) 6 3 (50%) (clinical)
TOTAL 30 27 (90%) 30 12 (40%)
TOTAL OF BOTH STUDIES 59 49 (88%) 58 23 (40%) These data demonstrate that the chimeric human antibody directed against LTA is opsonic and enhances survival against staphylococci. In addition, the
antibody promotes clearance of the staphylococci form the blood. Thus antibody to LTA provides prophylactic and therapeutic capabilities against staphylococcal infections and vaccines using LTA or peptide mimeotopes of LTA that induce anti- LTA antibodies would also have prophylactic capabilities.
Having now fully described the invention, it will be appreciated by those
skilled in the art that the invention can be performed within a range of equivalents and conditions without departing from the spirit and scope of the invention and without undue experimentation. In addition, while the invention has been described in light of certain embodiments and examples, the inventors believe that it is capable of further modifications. This application is intended to cover any variations, uses, or adaptions of the invention which follow the general principles set forth above.
The specification includes recitation to the literature and those literature references are herein specifically incorporated by reference.
The specification and examples are exemplary only with the particulars of the
claimed invention set forth as follows:
98/5
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: HENRY JACKSON FOUNDATION FOR THE ADVANCEMENT OF MILITARY MEDICINE
(ii) TITLE OF INVENTION: OPSONIC AND PROTECTIVE MONOCLONAL AND
CHIMERIC ANTIBODIES SPECIFIC FOR LIPOTEICHOIC ACID OF GRAM POSITIVE BACTERIA
(iii) NUMBER OF SEQUENCES: 105
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: FINNEGAN, HENDERSON, FARABOW, GARRETT &
DUNNER, LLP
(B) STREET: 1300 I Street, NW
(C) CITY: Washington
(D) STATE: DC
(E) COUNTRY : USA
(F) ZIP: 20005-3315
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentin Release #1.0, Version #1.30
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: PCT Unassigned
(B) FILING DATE: Concurrently Herewith
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Einaudi , Carol P.
(B) REGISTRATION NUMBER: 32,220
(C) REFERENCE/DOCKET NUMBER: 04995.0041-00304
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 202-408-4000
(B) TELEFAX: 202-408-4400 (2) INFORMATION FOR SEQ ID NO : 1 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS:
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 1 :
Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser Pro 1 5 10 15
(2) INFORMATION FOR SEQ ID NO : 2 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS:
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 2 :
Trp His Trp Arg His Arg lie Pro Leu Gin Leu Ala Ala Gly Arg 1 5 10 15
[ 2 ) INFORMATION FOR SEQ ID NO : 3 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 3 : TGAATTTTCT GTATGAGGTT T 21 (2) INFORMATION FOR SEQ ID NO : 4 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
lix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..30
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 4
GGG GCT CAT GCG GAT AGG GTT TAT GGG GCC 30
Gly Ala His Ala Asp Arg Val Tyr Gly Ala
1 5 10
(2) INFORMATION FOR SEQ ID NO : 5 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 10 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 5 :
Gly Ala His Ala Asp Arg Val Tyr Gly Ala 1 5 10
(2) INFORMATION FOR SEQ ID NO: 6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
!ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..30
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 6 :
GGG ANT CAT GCG GAT AGG GTT TAT GGG GCC 30
Gly Xaa His Ala Asp Arg Val Tyr Gly Ala
1 5 10
(2) INFORMATION FOR SEQ ID NO : 7 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 10 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 7
Gly Xaa His Ala Asp Arg Val Tyr Gly Ala 1 5 10
(2) INFORMATION FOR SEQ ID NO : 8 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 8 :
GGG GCT TGG CAT TGG CGT CAT CGT ATT CCT CTT CAG CTT GCT GCT GGT 48 Gly Ala Trp His Trp Arg His Arg lie Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
CGT GGG GCC 57
Arg Gly Ala
(2) INFORMATION FOR SEQ ID NO : 9 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9:
Gly Ala Trp His Trp Arg His Arg lie Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
Arg Gly Ala (2) INFORMATION FOR SEQ ID NO: 10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
[ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10:
GGG GCT CGT CGG CAT GGT AAT TTT TCT CAT TTT TTT CAT CGG TCG TTG 48 Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
ATT GGG GCC 57 lie Gly Ala
(2) INFORMATION FOR SEQ ID NO: 11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11:
Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
He Gly Ala (2) INFORMATION FOR SEQ ID NO: 12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 12:
GGG GCT TGG AAG GCT TTG TTT AGT CAT TCT TAT CGT CCT CGG GGT TCG 48 Gly Ala Trp Lys Ala Leu Phe Ser His Ser Tyr Arg Pro Arg Gly Ser 1 5 10 15
GCT GGG GCC 57
Ala Gly Ala
(2) INFORMATION FOR SEQ ID NO: 13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 13:
Gly Ala Trp Lys Ala Leu Phe Ser His Ser Tyr Arg Pro Arg Gly Ser 1 5 10 15
Ala Gly Ala (2) INFORMATION FOR SEQ ID NO: 14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
;ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14:
GGG GCT AGG CAT TGG CGT CAT CGT ATT CCT CTT CAG CTT GCT GCT GGT 48 Gly Ala Arg His Trp Arg His Arg He Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
CGT GGG GCC 57
Arg Gly Ala
(2) INFORMATION FOR SEQ ID NO: 15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(Xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15:
Gly Ala Arg His Trp Arg His Arg He Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
Arg Gly Ala (2) INFORMATION FOR SEQ ID NO: 16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16:
GGG GCT TGG CAT TGG CGT CAT CGT ATT CCT CTT CAG CTT GCT GCT GGT 48 Gly Ala Trp His Trp Arg His Arg He Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
CGT GGG GCC 57
Arg Gly Ala
(2) INFORMATION FOR SEQ ID NO: 17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17:
Gly Ala Trp His Trp Arg His Arg He Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
Arg Gly Ala (2) INFORMATION FOR SEQ ID NO: 18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
( i) SEQUENCE DESCRIPTION: SEQ ID NO: 18:
GGG GCT TGG CAT TGG CGT CAT CGT ATT CCT CTT CAG CTT GCT GCT GGT 48 Gly Ala Trp His Trp Arg His Arg He Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
CGT GGG GCC 57
Arg Gly Ala
(2) INFORMATION FOR SEQ ID NO: 19:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19:
Gly Ala Trp His Trp Arg His Arg He Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
Arg Gly Ala 9
(2) INFORMATION FOR SEQ ID NO: 20:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
lix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 20:
GGG GCT CAG GTG GCT GTT TTG TAT CCT CCT TTG GCT GAT GCT ACT GAG 48 Gly Ala Gin Val Ala Val Leu Tyr Pro Pro Leu Ala Asp Ala Thr Glu 1 5 10 15
CTT GGG GCC 57
Leu Gly Ala
(2) INFORMATION FOR SEQ ID NO: 21:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 21:
Gly Ala Gin Val Ala Val Leu Tyr Pro Pro Leu Ala Asp Ala Thr Glu 1 5 10 15
Leu Gly Ala (2) INFORMATION FOR SEQ ID NO: 22:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
!iχ) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 22:
GGG GCT CGT CGG CAT GGT AAT TTT TCT CAT TTT TTT CAT CGG TCG TTG 48 Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
ATT GGG GCC 57
He Gly Ala
(2) INFORMATION FOR SEQ ID NO: 23:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 23:
Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
He Gly Ala (2) INFORMATION FOR SEQ ID NO: 24:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 24:
GGG GCT CGT CGG CAT GGT AAT TTT TCT CAT TTT TTT CAT CGG TCG TTG 48 Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
ATT GGG GCC 57
He Gly Ala
(2) INFORMATION FOR SEQ ID NO: 25:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 25:
Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
He Gly Ala (2) INFORMATION FOR SEQ ID NO: 26:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 26:
GGG GCT TGG CGT ATG TAT TTT TCT CAT CGT CAT GCG CAT CTT CGT AGT 48 Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
CCT GGG GCC 57
Pro Gly Ala
(2) INFORMATION FOR SEQ ID NO: 27:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 27:
Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
Pro Gly Ala (2) INFORMATION FOR SEQ ID NO: 28:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 28:
GGG GCT TGG CGT ATG TAT TTT TCT CAT CGT CAT GCG CAT CTT CGT AGT 48 Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
CCT GGG GCC 57
Pro Gly Ala
(2) INFORMATION FOR SEQ ID NO: 29:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 29:
Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
Pro Gly Ala (2) INFORMATION FOR SEQ ID NO: 30:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 30:
GGG GCT TGG CGG AAG TAT TTT TCT TAT CAT CAT GCG CAT CTT TGT AGT 48 Gly Ala Trp Arg Lys Tyr Phe Ser Tyr His His Ala His Leu Cys Ser 1 5 10 15
CCT GGG GCC 57
Pro Gly Ala
(2) INFORMATION FOR SEQ ID NO: 31:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 31:
Gly Ala Trp Arg Lys Tyr Phe Ser Tyr His His Ala His Leu Cys Ser 1 5 10 15
Pro Gly Ala INFORMATION FOR SEQ ID NO: 32:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 32:
GGG GCT TGG CGT ATG TAT TTT TCT CAT CGT CAT GCG CAT CTT CGT AGT 48 Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
CCT GGG GCC 57
Pro Gly Ala
(2) INFORMATION FOR SEQ ID NO: 33:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 33:
Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
Pro Gly Ala (2) INFORMATION FOR SEQ ID NO: 34:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 34:
GGG GCT TGG CGT ATG TAT TTT TCT CAT CGT CAT GCG CAT CTT CGT AGT 48 Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
CCT GGG GCC 57
Pro Gly Ala
(2) INFORMATION FOR SEQ ID NO: 35:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 35:
Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
Pro Gly Ala (2) INFORMATION FOR SEQ ID NO: 36:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 36 :
GGG GCT TGG CGT ATG TAT TTT TCT CAT CGT CAT GCG CAT CTT CGT AGT 4 Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
CCT GGG GCC 5
Pro Gly Ala
(2) INFORMATION FOR SEQ ID NO: 37:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 37:
Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
Pro Gly Ala (2) INFORMATION FOR SEQ ID NO: 38:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 38 :
GGG GCT CGT CGG CAT GGT AAT TTT TCT CAT TTT TTT CAT CGG TCG TTG 48 Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
ATT GGG GCC 57
He Gly Ala
(2) INFORMATION FOR SEQ ID NO : 39 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
( i) SEQUENCE DESCRIPTION: SEQ ID NO: 39:
Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
He Gly Ala (2) INFORMATION FOR SEQ ID NO: 40:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 40:
GGG GCT TGG CAT TGG CGT CAT CGT ATT CCT CTT CAG CTT GCT GCT GGT 48 Gly Ala Trp His Trp Arg His Arg He Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
CGT GGG GCC 57
Arg Gly Ala
(2) INFORMATION FOR SEQ ID NO: 41:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 41:
Gly Ala Trp His Trp Arg His Arg He Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
Arg Gly Ala (2) INFORMATION FOR SEQ ID NO: 42:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 42:
GGG GCT CGT CGG CAT GGT AAT TTT TCT CAT TTT TTT CAT CGG TCG TTG 48 Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
ATT GGG GCC 57
He Gly Ala
(2) INFORMATION FOR SEQ ID NO: 43:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 43:
Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
He Gly Ala INFORMATION FOR SEQ ID NO: 44:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 44:
GGG GCT GAT TGG ATT ACT TTT CAT CGT CGT CAT CAT GAT CGT GTT CTT 48 Gly Ala Asp Trp He Thr Phe His Arg Arg His His Asp Arg Val Leu 1 5 10 15
TCT GGG GCC 57
Ser Gly Ala
(2) INFORMATION FOR SEQ ID NO: 45:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 45:
Gly Ala Asp Trp He Thr Phe His Arg Arg His His Asp Arg Val Leu 1 5 10 15
Ser Gly Ala (2) INFORMATION FOR SEQ ID NO: 46:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 46:
GGG GCT GGT TGG ATT ACT TTT CAT CGT CGT CAT CAT GAT CGT GTT CTT 48 Gly Ala Gly Trp He Thr Phe His Arg Arg His His Asp Arg Val Leu 1 5 10 15
TCT GGG GCC 57
Ser Gly Ala
(2) INFORMATION FOR SEQ ID NO: 47:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 47:
Gly Ala Gly Trp He Thr Phe His Arg Arg His His Asp Arg Val Leu 1 5 10 15
Ser Gly Ala (2) INFORMATION FOR SEQ ID NO: 48:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 48:
GGG GCT GGG AAG GCT ATG TTT AGT CAT TCT TAT CGT CAT CGG GGT TCG 48 Gly Ala Gly Lys Ala Met Phe Ser His Ser Tyr Arg His Arg Gly Ser 1 5 10 15
GCT GGG GCC 57
Ala Gly Ala
(2) INFORMATION FOR SEQ ID NO: 49:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 49:
Gly Ala Gly Lys Ala Met Phe Ser His Ser Tyr Arg His Arg Gly Ser 1 5 10 15
Ala Gly Ala (2) INFORMATION FOR SEQ ID NO: 50:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 50:
GGG GCT GAT TGG ATT ACT TTT CAT CGT CGT CAT CAT GAT CGT GTT CTT 48 Gly Ala Asp Trp He Thr Phe His Arg Arg His His Asp Arg Val Leu 1 5 10 15
TCT GGG GCC 57
Ser Gly Ala
(2) INFORMATION FOR SEQ ID NO: 51:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 51:
Gly Ala Asp Trp He Thr Phe His Arg Arg His His Asp Arg Val Leu 1 5 10 15
Ser Gly Ala (2) INFORMATION FOR SEQ ID NO: 52:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 52:
GGG GCT AGT CGT CAT ATG CTT GCT CGG TGG TCG CGT TTG CTT GCT GTT 48 Gly Ala Ser Arg His Met Leu Ala Arg Trp Ser Arg Leu Leu Ala Val 1 5 10 15
CCT GGG GCC 57
Pro Gly Ala
(2) INFORMATION FOR SEQ ID NO: 53:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 53:
Gly Ala Ser Arg His Met Leu Ala Arg Trp Ser Arg Leu Leu Ala Val 1 5 10 15
Pro Gly Ala (2) INFORMATION FOR SEQ ID NO: 54:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 54:
GGG GCT GGG AAG GCT ATG TTT AGT CAT TCT TAT CGT CAT CGG GGT TCG 48 Gly Ala Gly Lys Ala Met Phe Ser His Ser Tyr Arg His Arg Gly Ser 1 5 10 15
GCT GGG GCC 57
Ala Gly Ala
(2) INFORMATION FOR SEQ ID NO: 55:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 55:
Gly Ala Gly Lys Ala Met Phe Ser His Ser Tyr Arg His Arg Gly Ser 1 5 10 15
Ala Gly Ala (2) INFORMATION FOR SEQ ID NO: 56:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 56:
GGG GCT TGG CAT TGG CGT CAT CGT ATT CCT CTT CAG CTT GCT GCT GGT 48 Gly Ala Trp His Trp Arg His Arg He Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
CGT GGG GCC 57
Arg Gly Ala
(2) INFORMATION FOR SEQ ID NO: 57:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 57:
Gly Ala Trp His Trp Arg His Arg He Pro Leu Gin Leu Ala Ala Gly 1 5 10 15
Arg Gly Ala (2) INFORMATION FOR SEQ ID NO: 58:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 58:
GGG GCT CGT CGG CAT GGT AAT TTT TCT CAT TTT TTT CAT CGG TCG TTG 48 Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
ATT GGG GCC 57
He Gly Ala
(2) INFORMATION FOR SEQ ID NO: 59:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 59:
Gly Ala Arg Arg His Gly Asn Phe Ser His Phe Phe His Arg Ser Leu 1 5 10 15
He Gly Ala 9
(2) INFORMATION FOR SEQ ID NO: 60:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 60:
GGG GCT TGG AAG GCT TTG TTT AGT CAT TCT TAT CGT CCT CGG GGT TCG 48 Gly Ala Trp Lys Ala Leu Phe Ser His Ser Tyr Arg Pro Arg Gly Ser 1 5 10 15
GCT GGG GCC 57
Ala Gly Ala
(2) INFORMATION FOR SEQ ID NO: 61:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 61:
Gly Ala Trp Lys Ala Leu Phe Ser His Ser Tyr Arg Pro Arg Gly Ser 1 5 10 15
Ala Gly Ala (2) INFORMATION FOR SEQ ID NO: 62:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 62:
GGG GCT CAG GTG GCT GTT TTG TAT CCT CCT TTG GCT GAT GCT ACT GAG 48 Gly Ala Gin Val Ala Val Leu Tyr Pro Pro Leu Ala Asp Ala Thr Glu 1 5 10 15
CTT GGG GCC 57
Leu Gly Ala
(2) INFORMATION FOR SEQ ID NO: 63:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 63:
Gly Ala Gin Val Ala Val Leu Tyr Pro Pro Leu Ala Asp Ala Thr Glu 1 5 10 15
Leu Gly Ala (2) INFORMATION FOR SEQ ID NO: 64:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 64:
GGG GCT TGG CGT ATG TAT TTT TCT CAT CGT CAT GCG CAT CTT CGT AGT 48 Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
CCT GGG GCC 57
Pro Gly Ala
(2) INFORMATION FOR SEQ ID NO: 65:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 65:
Gly Ala Trp Arg Met Tyr Phe Ser His Arg His Ala His Leu Arg Ser 1 5 10 15
Pro Gly Ala 9
(2) INFORMATION FOR SEQ ID NO: 66:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..30
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 66:
GGG GCT CAT GCG GAT AGG GTT TAT GGG GCC 30
Gly Ala His Ala Asp Arg Val Tyr Gly Ala
1 5 10
(2) INFORMATION FOR SEQ ID NO: 67:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 10 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 67:
Gly Ala His Ala Asp Arg Val Tyr Gly Ala 1 5 10
(2) INFORMATION FOR SEQ ID NO: 68:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 45 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer" (Xi) SEQUENCE DESCRIPTION: SEQ ID NO: 68: ATTTCAGGCC CAGCCGGCCA TGGCCGARGT RMAGCTKSAK GAGWC 45
(2) INFORMATION FOR SEQ ID NO: 69:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 45 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 69: ATTTCAGGCC CAGCCGGCCA TGGCCGARGT YCARCTKCAR CARYC 45
(2) INFORMATION FOR SEQ ID NO: 70:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 45 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 70: ATTTCAGGCC CAGCCGGCCA TGGCCCAGGT GAAGCTKSTS GARTC 45
(2) INFORMATION FOR SEQ ID NO: 71:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 45 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer" (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 71: ATTTCAGGCC CAGCCGGCCA TGGCCGAVGT GMWGCTKGTG GAGWC 45
(2) INFORMATION FOR SEQ ID NO: 72:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 45 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 72: ATTTCAGGCC CAGCCGGCCA TGGCCCAGGT BCARCTKMAR SARTC 45
(2) INFORMATION FOR SEQ ID NO: 73:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 35 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 73: GCTGCCACCG CCACCTGMRG AGACDGTGAS TGARG 35
(2) INFORMATION FOR SEQ ID NO: 74:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 35 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer" (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 74: GCTGCCACCG CCACCTGMRG AGACDGTGAS MGTRG 35 (2) INFORMATION FOR SEQ ID NO: 75:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 35 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 75:
GCTGCCACCG CCACCTGMRG AGACDGTGAS CAGRG 35
(2) INFORMATION FOR SEQ ID NO: 76:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 32 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 76:
CCCGGGCCAC CATGGAGACA GACACACTCC TG 32
(2) INFORMATION FOR SEQ ID NO: 77:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 35 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 77: CCCGGGCCAC CATGGATTTT CAAGTGCAGA TTTTC 35 (2) INFORMATION FOR SEQ ID NO: 78:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 32 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 78: CCCGGGCCAC CATGGAGWCA CAKWCTCAGG TC 32 (2) INFORMATION FOR SEQ ID NO: 79:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 79:
CCCGGGCCAC CATGKCCCCW RCTCAGYTTC TKG 33
(2) INFORMATION FOR SEQ ID NO: 80:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 31 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 80 CCCGGGCACC ATGAAGTTGC CTGTTAGGCT G 31 (2) INFORMATION FOR SEQ ID NO: 81:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 81:
GCACCTCCAG ATGTTAACTG CTC 23
(2) INFORMATION FOR SEQ ID NO: 82:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 54 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 82: TAATATCGCG ACAGCTACAG GTGTCCACTC CCGAAGTGAT GCTGGTGGAG WCTG 54
(2) INFORMATION FOR SEQ ID NO: 83:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 83: TTATAGAATT CTGAGGAGAC GGTGAGTGAG 30
(2) INFORMATION FOR SEQ ID NO: 84: 98/57994
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 29 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 84: TTAGGCGATA TCGTTCTCTC CCAGTCTCC 29
(2) INFORMATION FOR SEQ ID NO: 85:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 46 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer'
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 85: GTAACCGTTC GAAAAGTGTA CTTACGTTTT ATTTCCAGCA TGGTCC 46
(2) INFORMATION FOR SEQ ID NO: 86:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 90 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..90 (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 86:
GAA GTG ATG CTG GTG GAG TCT GGT GGA GGA TTG GTG CAG CCT AAA GGG 48 Glu Val Met Leu Val Glu Ser Gly Gly Gly Leu Val Gin Pro Lys Gly 1 5 10 15
TCA TTG AAA CTC TCA TGT GCA GCC TCT GGA TTC ACC TTC AAT 90
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn 20 25 30
(2) INFORMATION FOR SEQ ID NO: 87:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 87:
Glu Val Met Leu Val Glu Ser Gly Gly Gly Leu Val Gin Pro Lys Gly 1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn 20 25 30
(2) INFORMATION FOR SEQ ID NO: 88:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 88:
AAC TAC GCC ATG AAT TGG GTC CGC CAG GCT CCA GGA AAG GGT TTG GAA 48 Asn Tyr Ala Met Asn Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu 1 5 10 15 98/57
TGG GTT GCT Trp Val Ala
(2) INFORMATION FOR SEQ ID NO: 89:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 89:
Asn Tyr Ala Met Asn Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu 1 5 10 15
Trp Val Ala
(2) INFORMATION FOR SEQ ID NO: 90:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 90:
CGC ATA AGA AGT AAA AGT AAT AAT TAT GCA ACA TTT TAT GCC GAT TCA 48 Arg He Arg Ser Lys Ser Asn Asn Tyr Ala Thr Phe Tyr Ala Asp Ser 1 5 10 15
GTG AAA GAC 57
Val Lys Asp (2) INFORMATION FOR SEQ ID NO: 91:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 91:
Arg He Arg Ser Lys Ser Asn Asn Tyr Ala Thr Phe Tyr Ala Asp Ser 1 5 10 15
Val Lys Asp
(2) INFORMATION FOR SEQ ID NO: 92:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 96 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..96
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 92:
AGG TTC ACC ATC TCC AGA GAT GAT TCA CAA AGC ATG CTC TAT CTG CAA 48 Arg Phe Thr He Ser Arg Asp Asp Ser Gin Ser Met Leu Tyr Leu Gin 1 5 10 15
ATG AAC AAC TTG AAA ACT GAG GAC ACA GCC ATG TAT TAC TGT GTG AGA 96 Met Asn Asn Leu Lys Thr Glu Asp Thr Ala Met Tyr Tyr Cys Val Arg 20 25 30 (2) INFORMATION FOR SEQ ID NO: 93:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 32 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 93:
Arg Phe Thr He Ser Arg Asp Asp Ser Gin Ser Met Leu Tyr Leu Gin 1 5 10 15
Met Asn Asn Leu Lys Thr Glu Asp Thr Ala Met Tyr Tyr Cys Val Arg 20 25 30
(2) INFORMATION FOR SEQ ID NO: 94:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 69 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..69
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 94:
CGG GGG GCT TCA GGG ATT GAC TAT GCT ATG GAC TAC TGG GGT CAA GGA 48 Arg Gly Ala Ser Gly He Asp Tyr Ala Met Asp Tyr Trp Gly Gin Gly 1 5 10 15
ACC TCA CTC ACC GTC TCC TCA 69
Thr Ser Leu Thr Val Ser Ser 20 (2) INFORMATION FOR SEQ ID NO: 95:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
( i) SEQUENCE DESCRIPTION: SEQ ID NO: 95:
Arg Gly Ala Ser Gly He Asp Tyr Ala Met Asp Tyr Trp Gly Gin Gly 1 5 10 15
Thr Ser Leu Thr Val Ser Ser 20
(2) INFORMATION FOR SEQ ID NO: 96:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 69 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..69
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 96:
CAA ATT GTT CTC TCC CAG TCT CCA GCA ATC CTG TCT GCA TCT CCA GGG 48 Gin He Val Leu Ser Gin Ser Pro Ala He Leu Ser Ala Ser Pro Gly 1 5 10 15
GAA AAG GTC ACA ATG ACT TGC 69
Glu Lys Val Thr Met Thr Cys 20 (2) INFORMATION FOR SEQ ID NO: 97:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 97:
Gin He Val Leu Ser Gin Ser Pro Ala He Leu Ser Ala Ser Pro Gly 1 5 10 15
Glu Lys Val Thr Met Thr Cys 20
(2) INFORMATION FOR SEQ ID NO: 98:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..30
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 98:
AGG GCC AGC TCA AGT GTA AAT TAC ATG CAC 30
Arg Ala Ser Ser Ser Val Asn Tyr Met His 1 5 10
(2) INFORMATION FOR SEQ ID NO: 99:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 10 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 99:
Arg Ala Ser Ser Ser Val Asn Tyr Met His 1 5 10
(2) INFORMATION FOR SEQ ID NO: 100:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 66 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..66
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 100:
TGG TAC CAG CAG AAG CCA GGA TCC TCC CCC AAA CCC TGG ATT TCT GCC 48 Trp Tyr Gin Gin Lys Pro Gly Ser Ser Pro Lys Pro Trp He Ser Ala 1 5 10 15
ACA TCC AAC CTG GCT TCT 66
Thr Ser Asn Leu Ala Ser 20
(2) INFORMATION FOR SEQ ID NO: 101:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 22 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 101:
Trp Tyr Gin Gin Lys Pro Gly Ser Ser Pro Lys Pro Trp He Ser Ala 1 5 10 15
Thr Ser Asn Leu Ala Ser 20 (2) INFORMATION FOR SEQ ID NO: 102:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 96 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..96
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 102:
GGA GTC CCT GCT CGC TTC AGT GGC AGT GGG TCT GGG ACC TCT TAC TCT 48 Gly Val Pro Ala Arg Phe Ser Gly Ser Gly Ser Gly Thr Ser Tyr Ser 1 5 10 15
CTC ACA ATC AGC AGA GTG GAG GCT GAA GAT GCT GCC ACT TAT TAC TGC 96 Leu Thr He Ser Arg Val Glu Ala Glu Asp Ala Ala Thr Tyr Tyr Cys 20 25 30
(2) INFORMATION FOR SEQ ID NO: 103:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 32 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 103:
Gly Val Pro Ala Arg Phe Ser Gly Ser Gly Ser Gly Thr Ser Tyr Ser 1 5 10 15
Leu Thr He Ser Arg Val Glu Ala Glu Asp Ala Ala Thr Tyr Tyr Cys 20 25 30 (2) INFORMATION FOR SEQ ID NO: 104:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..57
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 104:
CAG CAG TGG AGT AGT AAC CCA CCC ACG TTC GGA GGG GGG ACC ATG CTG 48 Gin Gin Trp Ser Ser Asn Pro Pro Thr Phe Gly Gly Gly Thr Met Leu 1 5 10 15
GAA ATA AGA 57
Glu He Arg
(2) INFORMATION FOR SEQ ID NO: 105:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 105:
Gin Gin Trp Ser Ser Asn Pro Pro Thr Phe Gly Gly Gly Thr Met Leu 1 5 10 15
Glu He Arg

Claims

We claim:
1. A monoclonal antibody to lipoteichoic acid of Gram positive bacteria, wherein the monoclonal antibody (a) binds to lipoteichoic acid at a level that is twice background or greater and (b) enhances the opsonization of Gram positive bacteria by 75% or more.
2. The monoclonal antibody of claim 1 wherein the antibody binds a peptide sequence selected from the group consisting of:
WRMYFSHRHAHLRSP (SEQ ID NO 1) and WHWRHRIPLQLAAGR (SEQ ID NO 2).
3. The monoclonal antibody of claim 1 wherein the antibody is a chimeric non- human/human antibody.
4. The chimeric antibody of claim 3 comprising at least part of a human immunoglobulin constant region and at least part of a non-human immunoglobulin variable region having specificity to lipoteichoic acid of Gram
positive bacteria.
5. A chimeric immunoglobulin chain comprising at least part of a human
immunoglobulin constant region and at least part of a non-human immunoglobulin variable region having specificity to lipoteichoic acid of Gram positive bacteria.
6. The chimeric immunoglobulin chain of claim 5 wherein the constant region is selected from the group consisting of IgG, IgA, and IgM.
7. The chimeric immunoglobulin chain of claim 5 wherein the chain is selected from the group consisting of a heavy chain and a light chain.
8. The chimeric immunoglobulin chain of claim 7 wherein the chain is a light chain is selected from the group consisting of a kappa chain and a lambda chain.
9. An antibody to lipoteichoic acid of Gram positive bacteria wherein the antibody (a) binds to lipoteichoic acid at a level that is twice background or greater; (b) enhances the opsonization of Gram positive bacteria by 75% or more; and (c) binds to a peptide sequence selected from the group consisting of:
WRMYFSHRHAHLRSP (SEQ ID NO 1) and
WHWRHRIPLQLAAGR (SEQ ID NO 2).
10. A pharmaceutical composition comprising the antibody of one of claims 1 or 9,
or fragments, regions, or derivatives thereof, and a pharmaceutically acceptable
carrier.
11. A protective monoclonal antibody to lipoteichoic acid of Gram positive bacteria, wherein the antibody enhances survival in a lethal animal model by 10% or more.
12. The protective monoclonal antibody of claim 11 , wherein the antibody binds to a peptide sequence selected from the group consisting of:
WRMYFSHRHAHLRSP (SEQ ID NO: 1) and WHWRHRIPLQLAAGR (SEQ ID NO: 2).
13. A pharmaceutical composition comprising the antibody of claim 11 , or fragments, regions, or derivatives thereof, and a pharmaceutically acceptable carrier.
14. A method for treating a patient having an infection caused by a Gram positive bacteria comprising administering to the patient a therapeutically effective amount of the pharmaceutical composition of claim 10 or 13.
15. A method for preventing infections caused by Gram positive infections in a
patient comprising administering to the patient a prophyiactically effective amount of the pharmaceutical composition of claim 10 or 13.
16. An lipoteichoic acid epitope peptide mimic comprising a peptide sequence
selected from the group consisting of: (a) WRMYFSHRHAHLRSP(SEQIDNOI)
(b) WHWRHRIPLQLAAGR (SEQ ID NO 2), and
(c) peptide sequences that are substantially homologous tq he sequences of (a) or (b).
17. A peptide encoded by the DNA of the variable region of the anti-lipoteichoic antibody of Figure 12 or a sequence that is at least 70% homologous to that DNA.
18. The peptide of claim 17 wherein the variable region on a chain is selected from the group consisting of the heavy chain and light chain.
19. The peptide of claim 17 wherein the DNA of the variable region encodes one or more of the Complementarity Determining Regions.
20. The peptide of claim 19 wherein the variable region is on a chain selected from the group consisting of the heavy chain and light chain.
21. A peptide characterized by amino acids corresponding to one or more of the Complementarity Determining Regions of the variable region of the anti- lipoteichoic antibody of Figure 12 or amino acids that are at least 70%
homologous to the Complementarity Determining Regions.
22. The peptide of claim 21 wherein the variable region is selected from the group consisting of the heavy chain and the light chain.
23. A pharmaceutical composition comprising the peptides of any of claims 16-22 and a pharmaceutically acceptable carrier.
24. A method for treating a patient having an infection caused by a Gram positive
bacteria comprising administering to the patient a therapeutically effective amount of the pharmaceutical composition of claim 23.
25. A method for preventing infections caused by Gram positive bacteria in a patient comprising administering to the patient a prophyiactically effective amount of the pharmaceutical composition of claim 23.
26. A vaccine for preventing infections caused by Gram positive bacteria comprising a lipoteichoic acid antigen and a pharmaceutically acceptable carrier.
27. The vaccine of claim 26 wherein the lipoteichoic acid antigen comprises the epitope of the antigen or an epitope mimic.
28. The vaccine of claim 26 wherein the epitope is a peptide sequence selected from the group consisting of:
(a) WRMYFSHRHAHLRSP(SEQIDNOI)
(b) WHWRHRIPLQLAAGR (SEQ ID NO 2), and
(c) peptide sequences that are substantially homologous to the sequences of (a) or (b).
29. An animal lethality test for determining the in vivo activity of a composition to treat or prevent infections by Gram positive bacteria comprising the steps of: a) administering a lipid emulsion to at least two groups of suckling rodents; b) injecting into one group the composition to be tested and injecting into the other group a control substance; c) administering through a catheter an amount of Gram positive bacteria sufficient to cause lethal sepsis;
d) leaving the catheter under the skin of the rodent ; and d) assessing the affect of administration of the composition on either or both bacteremia and survival, wherein compositions that either reduce bacteremia or enhance survival are
useful to treat or prevent infections by Gram positive bacteria.
30. The method of claim 29 wherein the composition to be tested is an antibody to lipoteichoic acid of Gram positive bacteria or fragments thereof.
31. The method of claim 29 wherein the Gram positive bacteria is selected from the group consisting of Staphylococcus epidermidis, Staphylococcus hemolyticus, Staphylococcus hominus, Staphylococcus aureus, Staphylococcus mutans, Staphylococcus faecalis, and Staphylococcus pyogenes.
EP98931278A 1997-06-16 1998-06-16 Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria Expired - Lifetime EP0986577B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP10182580A EP2357198A1 (en) 1997-06-16 1998-06-16 Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US4987197P 1997-06-16 1997-06-16
US498711P 1997-06-16
PCT/US1998/012402 WO1998057994A2 (en) 1997-06-16 1998-06-16 Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP10001171.7 Division-Into 2010-02-04
EP10182580.0 Division-Into 2010-09-29

Publications (2)

Publication Number Publication Date
EP0986577A2 true EP0986577A2 (en) 2000-03-22
EP0986577B1 EP0986577B1 (en) 2011-02-09

Family

ID=21962176

Family Applications (2)

Application Number Title Priority Date Filing Date
EP98931278A Expired - Lifetime EP0986577B1 (en) 1997-06-16 1998-06-16 Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
EP10182580A Withdrawn EP2357198A1 (en) 1997-06-16 1998-06-16 Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP10182580A Withdrawn EP2357198A1 (en) 1997-06-16 1998-06-16 Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Country Status (10)

Country Link
US (5) US6610293B1 (en)
EP (2) EP0986577B1 (en)
JP (2) JP4691225B2 (en)
AT (1) ATE497977T1 (en)
AU (1) AU8144098A (en)
CA (1) CA2293732A1 (en)
DE (1) DE69842129D1 (en)
DK (1) DK0986577T3 (en)
ES (1) ES2363689T3 (en)
WO (1) WO1998057994A2 (en)

Families Citing this family (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080139789A1 (en) * 1990-10-22 2008-06-12 Henry M. Jackson Foundation For The Advancement Of Military Medicine Isolated Broadly Reactive Opsonic Immunoglobulin for Treating a Pathogenic Coagulase-Negative Staphylococcus Infection
US20060235209A9 (en) * 1997-03-10 2006-10-19 Jin-An Jiao Use of anti-tissue factor antibodies for treating thromboses
US5986065A (en) * 1997-03-10 1999-11-16 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof
US7749498B2 (en) * 1997-03-10 2010-07-06 Genentech, Inc. Antibodies for inhibiting blood coagulation and methods of use thereof
US20030109680A1 (en) * 2001-11-21 2003-06-12 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof
US6610293B1 (en) * 1997-06-16 2003-08-26 The Henry M. Jackson Foundation For The Advancement Of Military Medicine Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US7250494B2 (en) * 1998-06-15 2007-07-31 Biosynexus Incorporated Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
US6824997B1 (en) 1998-09-18 2004-11-30 Binax, Inc. Process and materials for the rapid detection of streptococcus pneumoniae employing purified antigen-specific antibodies
US9134303B1 (en) 1998-08-25 2015-09-15 Alere Scarborough, Inc. ICT immunoassay for Legionella pneumophila serogroup 1 antigen employing affinity purified antibodies thereto
US20020092987A1 (en) * 1998-09-05 2002-07-18 Taehee Cho Photo detect device using quantum dots and materialization method thereof
DE19912706A1 (en) * 1999-03-05 2000-09-07 Petry Genmedics Gmbh Dr New staphylococcal DNA for dltABCD operons, useful e.g. for identifying antibacterials and agents that reduce bacterial resistance to antimicrobials
US6790661B1 (en) * 1999-07-16 2004-09-14 Verax Biomedical, Inc. System for detecting bacteria in blood, blood products, and fluids of tissues
US20020051793A1 (en) * 2000-09-12 2002-05-02 Drabick Joseph J. Lipoteichoic acid immunogenic compositions and methods of making and using thereof
US20030190705A1 (en) * 2001-10-29 2003-10-09 Sunol Molecular Corporation Method of humanizing immune system molecules
US20030224000A1 (en) * 2001-12-21 2003-12-04 Kokai-Kun John Fitzgerald Methods for blocking or alleviating staphylococcal nasal colonization by intranasal application of monoclonal antibodies
US20040052779A1 (en) * 2001-12-21 2004-03-18 Stinson Jeffrey R. Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
AU2012254925B2 (en) * 2001-12-21 2016-10-13 Biosynexus Incorporated Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US7169903B2 (en) * 2001-12-21 2007-01-30 Biosynexus Incorporated Multifunctional monoclonal antibodies directed to peptidoglycan of gram-positive bacteria
AU2002364740A1 (en) * 2001-12-21 2003-07-30 Biosynexus Incorporated Multifunctional monoclonal antibodies directed to peptidoglycan of gram-positive bacteria
CA2507711A1 (en) * 2002-12-02 2004-06-17 Biosynexus Incorporated Wall teichoic acid as a target for anti-staphylococcal therapies and vaccines
AU2004255553B2 (en) * 2003-06-19 2009-08-20 Genentech, Inc. Compositions and methods for treating coagulation related disorders
TWI333977B (en) 2003-09-18 2010-12-01 Symphogen As Method for linking sequences of interest
GB2408573A (en) * 2003-11-25 2005-06-01 Univ Leicester Assay for the interaction of L-Ficolin with lipoteichoic acid
CA2580103C (en) 2004-09-22 2021-11-16 Glaxosmithkline Biologicals S.A. Immunogenic composition
US20060188526A1 (en) * 2005-02-24 2006-08-24 Cunnion Kenji M Method for enhancing the immune response to Staphylococcus aureus infection
WO2007008904A2 (en) 2005-07-08 2007-01-18 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Targeting poly-gamma-glutamic acid to treat staphylococcus epidermidis and related infections
EP1998802A2 (en) 2006-03-30 2008-12-10 GlaxoSmithKline Biologicals S.A. Immunogenic composition
NZ594945A (en) * 2006-06-06 2013-03-28 Crucell Holland Bv Human binding molecules having killing activity against enterococci and staphylococcus aureus and uses thereof
US7960518B2 (en) * 2006-06-06 2011-06-14 Crucell Holland B.V. Human binding molecules having killing activity against enterococci and uses thereof
AU2012268821B2 (en) * 2006-06-06 2016-06-02 Janssen Vaccines & Prevention B.V. Human binding molecules having killing activity against staphylococci and uses thereof
DK2395018T3 (en) * 2006-06-06 2016-04-25 Crucell Holland Bv HUMAN BINDING MOLECULES with killer activity against staphylococci and uses thereof
US8080645B2 (en) * 2007-10-01 2011-12-20 Longhorn Vaccines & Diagnostics Llc Biological specimen collection/transport compositions and methods
US8097419B2 (en) 2006-09-12 2012-01-17 Longhorn Vaccines & Diagnostics Llc Compositions and method for rapid, real-time detection of influenza A virus (H1N1) swine 2009
US9481912B2 (en) 2006-09-12 2016-11-01 Longhorn Vaccines And Diagnostics, Llc Compositions and methods for detecting and identifying nucleic acid sequences in biological samples
US8652782B2 (en) 2006-09-12 2014-02-18 Longhorn Vaccines & Diagnostics, Llc Compositions and methods for detecting, identifying and quantitating mycobacterial-specific nucleic acids
CA2677771A1 (en) * 2007-03-01 2008-09-04 Symphogen A/S Method for cloning cognate antibodies
US20100166772A1 (en) 2007-05-31 2010-07-01 Anderson Annaliesa S ANTIGEN-BINDING PROTEINS TARGETING S. AUREUS ORF0657n
US9683256B2 (en) 2007-10-01 2017-06-20 Longhorn Vaccines And Diagnostics, Llc Biological specimen collection and transport system
US11041215B2 (en) 2007-08-24 2021-06-22 Longhorn Vaccines And Diagnostics, Llc PCR ready compositions and methods for detecting and identifying nucleic acid sequences
CA2697373C (en) 2007-08-27 2019-05-21 Longhorn Vaccines & Diagnostics, Llc Immunogenic compositions and methods
US10004799B2 (en) 2007-08-27 2018-06-26 Longhorn Vaccines And Diagnostics, Llc Composite antigenic sequences and vaccines
KR101661946B1 (en) * 2007-08-31 2016-10-05 유니버시티 오브 시카고 Methods and compositions related to immunizing against staphylococcal lung diseases and conditions
US9181329B2 (en) 2007-08-31 2015-11-10 The University Of Chicago Methods and compositions related to immunizing against Staphylococcal lung diseases and conditions
US11041216B2 (en) 2007-10-01 2021-06-22 Longhorn Vaccines And Diagnostics, Llc Compositions and methods for detecting and quantifying nucleic acid sequences in blood samples
WO2009085355A2 (en) 2007-10-01 2009-07-09 Longhorn Vaccines & Diagnostics Llc Biological specimen collection and transport system and methods of use
US8758765B2 (en) * 2008-07-29 2014-06-24 The University Of Chicago Compositions and methods related to Staphylococcal bacterium proteins
US20100082438A1 (en) * 2008-10-01 2010-04-01 Ronnie Jack Garmon Methods and systems for customer performance scoring
WO2010085590A1 (en) 2009-01-23 2010-07-29 Biosynexus Incorporated Opsonic and protective antibodies specific for lipoteichoic acid gram positive bacteria
PL3281947T3 (en) 2009-04-03 2020-07-27 The University Of Chicago Compositions and methods related to protein a (spa) variants
PL2454284T3 (en) 2009-07-15 2018-09-28 Aimm Therapeutics B.V. Gram-positive bacteria specific binding compounds
CA2772240C (en) 2009-08-17 2017-12-05 Tracon Pharmaceuticals, Inc. Combination therapy of cancer with anti-endoglin antibodies and anti-vegf agents
US8221753B2 (en) 2009-09-30 2012-07-17 Tracon Pharmaceuticals, Inc. Endoglin antibodies
WO2011123452A1 (en) * 2010-03-29 2011-10-06 Biosynexus Incorporated Compositions and methods for prophylactic and therapeutic treatment of infection
JP2013523818A (en) 2010-04-05 2013-06-17 ザ・ユニバーシティー・オブ・シカゴ Compositions and methods relating to protein A (SpA) antibodies as enhancers of immune responses
JP6002128B2 (en) 2010-07-02 2016-10-05 ザ・ユニバーシティ・オブ・シカゴThe University Of Chicago Compositions and methods related to protein A (SpA) variants
WO2012034067A1 (en) 2010-09-09 2012-03-15 The University Of Chicago Methods and compositions involving protective staphylococcal antigens
US8945588B2 (en) 2011-05-06 2015-02-03 The University Of Chicago Methods and compositions involving protective staphylococcal antigens, such as EBH polypeptides
CN103906535B (en) 2011-08-15 2017-07-14 芝加哥大学 The composition related to the antibody of staphylococcal protein A and method
WO2013071409A1 (en) 2011-11-18 2013-05-23 National Research Council Of Canada (Nrc) Clostridium difficile lipoteichoic acid and uses thereof
CA2863083C (en) 2012-01-26 2023-09-19 Longhorn Vaccines And Diagnostics, Llc Composite antigenic sequences and vaccines
DK2844275T3 (en) 2012-04-26 2020-07-13 Univ Chicago Staphylococcal coagulase antigens and methods of using them
NZ702282A (en) 2012-04-26 2016-07-29 Univ Chicago Compositions and methods related to antibodies that neutralize coagulase activity during staphylococcus aureus disease
CA2879272A1 (en) 2012-07-16 2014-01-23 Robert G.K. DONALD Saccharides and uses thereof
UA115789C2 (en) 2012-09-05 2017-12-26 Трейкон Фармасутікалз, Інк. Antibody formulations and uses thereof
GB201310008D0 (en) 2013-06-05 2013-07-17 Glaxosmithkline Biolog Sa Immunogenic composition for use in therapy
US9814766B2 (en) 2013-12-28 2017-11-14 Longhorn Vaccines And Diagnostics, Llc Multimodal antimicrobial therapy
US9926375B2 (en) 2014-11-12 2018-03-27 Tracon Pharmaceuticals, Inc. Anti-endoglin antibodies and uses thereof
JP2017537084A (en) 2014-11-12 2017-12-14 トラコン ファーマシューティカルズ、インコーポレイテッド Anti-endoglin antibodies and uses thereof
AU2015359503B2 (en) 2014-12-10 2019-05-09 Glaxosmithkline Biologicals Sa Method of treatment
EP3294448A4 (en) 2015-05-14 2018-12-12 Longhorn Vaccines and Diagnostics, LLC Rapid methods for the extraction of nucleic acids from biological samples
JP7117244B2 (en) 2016-02-12 2022-08-12 ザ・ユニバーシティ・オブ・シカゴ Compositions and methods related to antibodies that neutralize coagulase activity during Staphylococcus aureus disease
KR20220107166A (en) 2019-10-02 2022-08-02 얀센 백신스 앤드 프리벤션 비.브이. Staphylococcus Peptides and Methods of Use

Family Cites Families (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1499035A (en) 1975-04-10 1978-01-25 Ts Nii Gematologii I Perelivan Antistaphylococcus human immune globulin and method of preparing same
JPS5452794A (en) 1977-09-30 1979-04-25 Kousaku Yoshida Extracting of polysacchride from capusle containing epidermis staphylococus
US4732757A (en) 1978-02-06 1988-03-22 Stolle Research And Development Corporation Prevention and treatment of rheumatoid arthritis
USRE33565E (en) * 1978-02-06 1991-04-02 Stolle Research And Development Corporation Prevention and treatment of rheumatioid arthritis
US4235869A (en) * 1978-05-16 1980-11-25 Syva Company Assay employing a labeled Fab-fragment ligand complex
DE2901822A1 (en) * 1979-01-18 1980-07-31 Biotest Serum Institut Gmbh METHOD FOR THE PRODUCTION OF AN IMMUNOGLOBULIN SOLUTION SUITABLE FOR THE INTRAVENOUS APPLICATION THAT IGM CONTAINS IN A CONCENTRATED FORM
FR2475900A1 (en) 1980-02-20 1981-08-21 Fabre Sa Pierre VACCINE COMPLEX CONTAINING A SPECIFIC ANTIGEN AND VACCINE CONTAINING SAME
US4425330A (en) 1981-05-20 1984-01-10 Cornell Research Foundation, Inc. Bovine mastitis vaccine and method for detecting efficacy thereof
US4744982A (en) * 1982-08-24 1988-05-17 Hunter Kenneth W Human monoclonal antibody reactive with polyribosylribitol phosphate
US4954449A (en) * 1982-08-24 1990-09-04 The United States Of America As Represented By The Secretary Of The Army Human monoclonal antibody reactive with polyribosylribitol phosphate
US4482483A (en) 1983-04-06 1984-11-13 Armour Pharmceutical Company Composition of intravenous immune globulin
US4761283A (en) * 1983-07-05 1988-08-02 The University Of Rochester Immunogenic conjugates
US4719290A (en) 1983-09-02 1988-01-12 Armour Pharmaceutical Corporation Composition of intravenous immune globulin
US4596769A (en) * 1984-03-05 1986-06-24 Temple University Monoclonal antibodies to peptidoglycan and methods of preparing same
US5043267A (en) * 1984-05-18 1991-08-27 E. I. Du Pont De Nemours And Company Method for rapid detection of bacterial and fungal infection
NZ214503A (en) 1984-12-20 1990-02-26 Merck & Co Inc Covalently-modified neutral bacterial polysaccharides, stable covalent conjugates of such polysaccharides and immunogenic proteins, and methods of preparing such polysaccharides and conjugates
US4888279A (en) * 1984-12-21 1989-12-19 Thomas Jefferson University Novel immunosorbent assays employing antibiotic keying agents
DE3516119A1 (en) * 1985-05-04 1986-11-06 Biotest Pharma GmbH, 6000 Frankfurt POLYVALENT HYPERIMMIMMLOBULIN PREPARATE
FR2581877B1 (en) 1985-05-14 1987-12-18 Louvain Universite Catholique CONJUGATE CONSISTING OF A WALL ADHESIN OF S. MUTANS, OF PROTEIN NATURE AND OF A POLYSACCHARIDE OF S. MUTANS, ITS PREPARATION AND ITS USE IN PARTICULAR IN CARIES VACCINES
CA1341235C (en) 1987-07-24 2001-05-22 Randy R. Robinson Modular assembly of antibody genes, antibodies prepared thereby and use
FR2619122B1 (en) 1987-08-03 1990-03-09 Pasteur Institut PROCESS FOR OBTAINING CAPSULAR POLYOSIDES OF STAPHYLOCOCCS, POLYOSIDES OBTAINED, APPLICATIONS OF SUCH POLYOSIDES AND STRAINS FOR IMPLEMENTING THE PROCESS
US5034515A (en) 1987-09-22 1991-07-23 Wisconsin Alumni Research Foundation Staphylococcal fibronectin receptor, monoclonal antibodies thereto and methods of use
JP2666212B2 (en) * 1988-03-07 1997-10-22 鐘紡株式会社 Antibody and method for producing anti-caries agent containing the same as an active ingredient
US5055455A (en) 1988-09-28 1991-10-08 Brigham And Women's Hospital Capsular polysaccharide adhesin antigen, preparation, purification and use
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
SE8901687D0 (en) 1989-05-11 1989-05-11 Alfa Laval Agri Int FIBRONECTIN BINDING PROTEIN AS WELL AS IT'S PREPARATION
US5069896A (en) * 1989-06-01 1991-12-03 The Gillette Company Malodor inhibiting skin deodorant composition comprising a monoclonal antibody and method of deodorizing
US5440014A (en) 1990-08-10 1995-08-08 H+E,Uml/Oo/ K; Magnus Fibronectin binding peptide
US5153312A (en) 1990-09-28 1992-10-06 American Cyanamid Company Oligosaccharide conjugate vaccines
US5571511A (en) * 1990-10-22 1996-11-05 The U.S. Government Broadly reactive opsonic antibodies that react with common staphylococcal antigens
US5955074A (en) * 1990-10-22 1999-09-21 Henry M. Jackson Foundation For The Advancement Of Military Medicine Directed human immune globulin for the prevention and treatment of staphylococcal infections
USRE37525E1 (en) * 1991-05-01 2002-01-22 Henry M. Jackson Foundation Method for treating infectious respiratory diseases
WO1993006213A1 (en) * 1991-09-23 1993-04-01 Medical Research Council Production of chimeric antibodies - a combinatorial approach
ES2198405T3 (en) 1991-11-22 2004-02-01 Nabi Biopharmaceuticals TYPE I SURFACE ANTIGENS ASSOCIATED WITH STAPHYLOCOCCUS EPIDERMIS.
CA2117480A1 (en) 1992-02-25 1993-09-02 Gerald W. Fischer Directed human immune globulin for the prevention and treatment of staphylococcal infections
AU3805893A (en) * 1992-03-19 1993-10-21 U.S. Government, As Represented By The Secretary Of The Army Broadly reactive opsonic antibodies that react with common staphylococcal antigens
ES2091684T3 (en) 1992-11-13 1996-11-01 Idec Pharma Corp THERAPEUTIC APPLICATION OF CHEMICAL AND RADIO-MARKED ANTIBODIES AGAINST THE RESTRICTED DIFFERENTIATION ANTIGEN OF HUMAN B-LYMPHOCYTES FOR THE TREATMENT OF B-CELL LYMPHOMA.
US5545721A (en) 1992-12-21 1996-08-13 Ophidian Pharmaceuticals, Inc. Conjugates for the prevention and treatment of sepsis
EP0680337A4 (en) * 1993-01-12 1997-07-30 Anthony George Gristina Methods and compositions for the direct concentrated delivery of passive immunity.
US5955078A (en) 1993-02-05 1999-09-21 Smithkline Beecham P.L.C. Fibronectin binding protein polypeptides
US6080407A (en) * 1993-05-17 2000-06-27 The Picower Institute For Medical Research Diagnostic assays for MIF
US5354654A (en) 1993-07-16 1994-10-11 The United States Of America As Represented By The Secretary Of The Navy Lyophilized ligand-receptor complexes for assays and sensors
US5624904A (en) 1993-11-17 1997-04-29 Massachusetts Institute Of Technology Method for treating gram positive septicemia
US5585098A (en) * 1993-11-23 1996-12-17 Ovimmune, Inc. Oral administration of chicken yolk immunoglobulins to lower somatic cell count in the milk of lactating ruminants
US5538733A (en) 1994-07-07 1996-07-23 Willmar Poultry Company, Inc. Method of priming an immune response in a one-day old animal
DE69534601T2 (en) * 1994-09-16 2006-08-03 The Scripps Research Institute, La Jolla USE OF ANTIBODIES TO PREVENT EFFECTS CAUSED BY GRAMPOSITIVE AND MYKO BACTERIA
JP4160633B2 (en) 1994-09-21 2008-10-01 ヘンリー エム.ジャクソン ファウンデーション フォー ザ アドバンスメント オブ ミリタリー メディスン Opsonizing antibodies that react extensively with common staphylococcal antigens
FR2729960A1 (en) 1995-01-30 1996-08-02 Bio Merieux TOXOPLASMA GONDII MIMOTOPE POLYPEPTIDES AND APPLICATIONS
EP0807185B9 (en) * 1995-01-30 2002-12-11 Lunamed AG Antitumor preparations containing a lipoteichoic acid from steptococcus
AU6094696A (en) * 1995-06-05 1996-12-24 Bionebraska, Inc. Lead binding polypeptides and nucleotides coding therefor
MA24512A1 (en) 1996-01-17 1998-12-31 Univ Vermont And State Agrienl PROCESS FOR THE PREPARATION OF ANTICOAGULATING AGENTS USEFUL IN THE TREATMENT OF THROMBOSIS
EP0950068B1 (en) * 1996-05-16 2005-11-09 THE TEXAS A&M UNIVERSITY SYSTEM Collagen binding protein compositions and methods of use
GB9614274D0 (en) * 1996-07-06 1996-09-04 Univ Manchester Treatment and diagnosis of infections of gram positive cocci
US5770208A (en) * 1996-09-11 1998-06-23 Nabi Staphylococcus aureus B-linked hexosamine antigen
US6294177B1 (en) * 1996-09-11 2001-09-25 Nabi Staphylococcus aureus antigen-containing whole cell vaccine
US6610293B1 (en) * 1997-06-16 2003-08-26 The Henry M. Jackson Foundation For The Advancement Of Military Medicine Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US6248329B1 (en) * 1998-06-01 2001-06-19 Ramaswamy Chandrashekar Parasitic helminth cuticlin nucleic acid molecules and uses thereof
US7250494B2 (en) 1998-06-15 2007-07-31 Biosynexus Incorporated Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
US6487198B1 (en) 1998-06-16 2002-11-26 Mci Communications Corporation Method and system for unloading T1 payloads from ATM cells
DE69940371D1 (en) * 1998-09-14 2009-03-19 Nabi Biopharmaceuticals Rockvi BETA-GLUCAN CONTAINING COMPOSITIONS AND SPECIFIC IMMUNOGLOBULINS
CN1355690A (en) * 1999-04-16 2002-06-26 奥赛尔股份有限公司 Method for improving half-life of soluble viral-specific ligands on mucosal membranes
US20040052779A1 (en) * 2001-12-21 2004-03-18 Stinson Jeffrey R. Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9857994A2 *

Also Published As

Publication number Publication date
AU8144098A (en) 1999-01-04
ES2363689T3 (en) 2011-08-11
EP2357198A1 (en) 2011-08-17
JP2002503966A (en) 2002-02-05
JP4691225B2 (en) 2011-06-01
CA2293732A1 (en) 1998-12-23
DE69842129D1 (en) 2011-03-24
US6939543B2 (en) 2005-09-06
DK0986577T3 (en) 2011-05-23
EP0986577B1 (en) 2011-02-09
US20060002939A1 (en) 2006-01-05
WO1998057994A2 (en) 1998-12-23
US7511122B2 (en) 2009-03-31
US7884198B2 (en) 2011-02-08
JP2011026327A (en) 2011-02-10
WO1998057994A3 (en) 1999-03-25
ATE497977T1 (en) 2011-02-15
US8372958B2 (en) 2013-02-12
US20020082395A1 (en) 2002-06-27
US20040013673A1 (en) 2004-01-22
US20100221822A1 (en) 2010-09-02
US6610293B1 (en) 2003-08-26

Similar Documents

Publication Publication Date Title
EP0986577B1 (en) Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US7777017B2 (en) Nucleic acids encoding opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
AU2009202762B2 (en) Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
CA2303202C (en) Amino acid sequences for therapeutical and prophylactic applications to diseases due to clostridium difficile toxins
EP1690875A1 (en) Human monoclonal antibody specific for lipopolysaccharides (LPS) of the pseudomonas aeruginosa IATS O11 serotype
JP2005514053A6 (en) Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram-positive bacteria
CA2525657C (en) Human monoclonal antibody specific for lipopolysaccharides (lps) of serotype iats o6 of pseudomonas aeruginosa
AU2010201603B2 (en) Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
Borghesi et al. Increase of cross (auto)-reactive antibodies after immunization in aged mice: a cellular and molecular study.
AU2012201369A1 (en) Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
AU2012254925B2 (en) Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20000117

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 16/46 20060101ALI20100831BHEP

Ipc: C12N 5/20 20060101ALI20100831BHEP

Ipc: A61P 31/04 20060101ALI20100831BHEP

Ipc: A61K 39/40 20060101ALI20100831BHEP

Ipc: C07K 16/12 20060101ALI20100831BHEP

Ipc: C07K 16/00 20060101AFI20100831BHEP

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REF Corresponds to:

Ref document number: 69842129

Country of ref document: DE

Date of ref document: 20110324

Kind code of ref document: P

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 69842129

Country of ref document: DE

Effective date: 20110324

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110609

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110209

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110510

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: KIRKER & CIE S.A.

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2363689

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20110811

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110209

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110209

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110209

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20111110

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 69842129

Country of ref document: DE

Effective date: 20111110

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20120626

Year of fee payment: 15

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20110630

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20110616

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20130627

Year of fee payment: 16

Ref country code: CH

Payment date: 20130627

Year of fee payment: 16

Ref country code: DE

Payment date: 20130627

Year of fee payment: 16

Ref country code: IE

Payment date: 20130625

Year of fee payment: 16

Ref country code: DK

Payment date: 20130625

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20130625

Year of fee payment: 16

Ref country code: FR

Payment date: 20130702

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20130627

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20130626

Year of fee payment: 16

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 69842129

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20140630

REG Reference to a national code

Ref country code: NL

Ref legal event code: V1

Effective date: 20150101

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20140616

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20150227

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150101

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 69842129

Country of ref document: DE

Effective date: 20150101

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140630

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140630

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140616

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140616

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150101

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140616

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140630

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20150729

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140630

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140617

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140630